WO2006044762A2 - Tetrahydro-5h-pyrimido[4,5-d]azepine derivatives useful for the treatment of diseases associated with the 5-ht2c receptor - Google Patents

Tetrahydro-5h-pyrimido[4,5-d]azepine derivatives useful for the treatment of diseases associated with the 5-ht2c receptor Download PDF

Info

Publication number
WO2006044762A2
WO2006044762A2 PCT/US2005/037193 US2005037193W WO2006044762A2 WO 2006044762 A2 WO2006044762 A2 WO 2006044762A2 US 2005037193 W US2005037193 W US 2005037193W WO 2006044762 A2 WO2006044762 A2 WO 2006044762A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
optionally substituted
disorders
compound
inhibitors
Prior art date
Application number
PCT/US2005/037193
Other languages
French (fr)
Other versions
WO2006044762A3 (en
Inventor
Ann-Marie Campbell
Philip Coish
Stephen O'connor
Evelyn Sibley
Harold Wright
Ming Wang
Donald Bierer
Original Assignee
Bayer Pharmaceuticals Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bayer Pharmaceuticals Corporation filed Critical Bayer Pharmaceuticals Corporation
Publication of WO2006044762A2 publication Critical patent/WO2006044762A2/en
Publication of WO2006044762A3 publication Critical patent/WO2006044762A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the invention relates to tetrahydro-5H -pyrimido[4,5-d]azepine compounds of Formula (I) that are useful in the treatment or prevention of diseases and/or behaviors that involve the 5-HT 2C receptor.
  • the compounds of the present invention may also be used for the treatment or prevention of diseases and/or disorders including obesity, obesity-related disorders such as diabetes, feeding behavior disorders, eating disorders such as bulimia and anorexia nervosa, and premenstrual tension.
  • Obesity which is an excess of body fat relative to lean body mass, is a chronic disease that is highly prevalent in modem society. It is associated not only with a social stigma, but also with decreased life span and numerous medical problems, including adverse psychological development, coronary artery disease, hypertension, stroke, diabetes, hyperlipidaemia, and some cancers (see, e.g., Nishina, et al., Metab. 43:554- 558, 1994; Grundy and Barnett, Dis. Mon. 36:641-731 , 1990; Rissanen, et al., British Medical Journal, 301 :835-837, 1990).
  • Obesity is considered a major medical problem largely because it is a factor for a number of other diseases. For example, obesity is correlated with a much higher incidence of type 2 diabetes (non-insulin dependent diabetes mellitus, NIDDM), hypertension, hyperlipidemia, myocardial infarction, cancers, gallbladder disease, respiratory disease, gout, arthritis, and dermatological disease. In addition, obese individuals have a higher incidence of mortality at a younger age as compared to their leaner counterparts.
  • type 2 diabetes non-insulin dependent diabetes mellitus, NIDDM
  • hypertension hyperlipidemia
  • myocardial infarction cancers
  • cancers gallbladder disease
  • respiratory disease gout
  • arthritis gout
  • Serotonin (5-HT) has been implicated in the regulation of feeding behavior.
  • Drugs which increase the concentration of 5-HT in the synaptic cleft by increasing 5-HT release and/or inhibiting re-uptake of the transmitter are effective long-term treatments for obesity, albeit with significant side-effects.
  • Serotonin produces physiological effects by acting on a heterogeneous family of receptors.
  • the lack of selective agonists and antagonists for the individual subtypes of serotonin receptors has prevented a complete characterization of the physiological role of each receptor subtype.
  • 5-HT 1A , 5-HT 1B , 5-HT 2A , 5-HT 2B , 5-HT 6 , and 5-HT 2C has been demonstrated to elicit effects on food intake.
  • activation of both 5-HT 2A and 5-HT 2C receptors decrease food intake.
  • 5-HT 2A receptor agonists are thought to decrease food intake by disrupting the ability of the animal to feed
  • the 5-HT 2C receptor has been implicated in the regulation of satiety.
  • 5- HT 2C receptors are localized to the hypothalamus and the brainstem, two brain regions known to play a critical role in the modulation of food intake.
  • 5-HT 2C receptor agonists produce a decrease in food intake which is associated with the least likely potential for side effects.
  • agonists that are selective for this receptor would have superior properties with respect to other known appetite suppressants, such as serotonin/noradrenaline re-uptake inhibitors and/or releasers, which can lead to hypertension and/or cardiac valve defects.
  • Non-selective agonists/partial agonists (e.g., mCPP, TFMPP) of the 5-HT 2C receptor have been shown to reduce food intake in rats and to accelerate the appearance of the behavioral satiety sequence.
  • hypophagic effects of mCPP are antagonized by the selective 5-HT 2C receptor antagonist, SB-242084.
  • Recent findings from studies in normal human volunteers and obese subjects administered mCPP have also shown decreases in food intake. A single injection of mCPP decreased food intake in female volunteers and subchronic treatment for a 14 day period decreased the appetite and body weight of obese male and female subjects.
  • the compounds of the present invention interact with the 5-HT 2C receptor.
  • these compounds may be useful for treatment or prevention of diseases and/or behaviors associated with the 5-HT 2C receptor.
  • the compounds of the present invention may also be used for the treatment and/or prevention of obesity, obesity-related disorders such as diabetes, feeding behavior, eating disorders such as bulimia and anorexia nervosa, and premenstrual tension.
  • other diseases and/or disorders such as central nervous disorders, depression, anxiety disorders, obsessive-compulsive disorders, sleep disorders, sexual dysfunction, psychoses, migraine, schizophrenia, drug or alcohol addiction, and chronic fatigue syndrome may also be treated and/or prevented by the compounds of the present invention.
  • the invention relates to a compound of Formula (I)
  • phenylpyrrolidin-1 -yl optionally substituted on the phenyl ring with up to two groups selected from halo, (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, or CF 3 ,
  • phenylpiperazin-1 -yl optionally substituted on the phenyl ring with up to two groups selected from (C 1 -C 3 )alkyl, halo, CF 3 , or (C 1 -C 3 )alkoxy,
  • R 5 is H or (C 1 -C 3 )alkyl
  • R 6 is (C 1 -C 3 )alkyl optionally substituted with
  • phenyl optionally substituted with up to two groups selected from (C 1 -C 3 )alkoxy, or benzodioxan-6-yl, or
  • phenyl optionally substituted with up to two groups selected from CF 3 , halo, (C 1 -C 3 )alkyl, NO 2 , (C 1 -C 3 )alkoxy, or N[(C 1 -C 3 )alkyl] 2 ,
  • R 3 is H or (C 1 -C 3 )alkyl
  • R 4 is H, methyl, -C(O)O-R 8 , or -C(O)R 9 ;
  • R 8 is (C 1 -C 4 )alkyl or phenyl optionally substituted with halo, (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, or CF 3 , and
  • R 9 is (C 1 -C 4 )alkyl or phenyl optionally substituted with halo, (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, or CF 3 ;
  • (C 1 -C 3 )alkyl mean a linear or branched saturated hydrocarbon groups having from 1 to about 3, from 1 to about 4, and from 1 to about 6 carbon atoms, respectively.
  • Such groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, pentyl, hexyl, and the like.
  • (C 1 -C 3 )alkoxy means a linear or branched saturated hydrocarbon group having from about 1 to about 3 carbon atoms, said group being attached to an oxygen atom.
  • the oxygen atom is the atom through which the alkoxy substituent is attached to the rest of the molecule.
  • groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, and the like.
  • halo means F, Br, Cl, and I.
  • (C 3 -C 6 )cycloalkyl means a saturated monocyclic alkyl group of from 3 to about 6 carbon atoms and includes, but is not limited to, such groups as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
  • optionally substituted means that the moiety so modified may be unsubstituted or substituted with the identified substituent(s). When any moiety is described as being substituted, it can have one or more of the indicated substituents that may be located at any available position on the moiety. When there are two or more substituents on any moiety, each substituent may be defined independently of any other substituent and may, accordingly, be the same or different.
  • Each substituent may replace any H atom on the moiety so modified as long as the replacement is chemically possible and chemically stable.
  • each substituent is chosen independently of any other substituent and can, accordingly, be the same or different.
  • the compounds of Formula (I) may contain one or more asymmetric centers, depending upon the location and nature of the various substituents desired.
  • Asymmetric carbon atoms may be present in the (R) or (S) configuration.
  • Preferred isomers are those with the absolute configuration which produces the compound of Formula (I) with the more desirable biological activity.
  • asymmetry may also be present due to restricted rotation about a given bond, for example, the central bond adjoining two aromatic rings of the specified compounds.
  • Substituents on a ring may also be present in either cis or trans form, and a substituent on a double bond may be present in either Z or E form.
  • Representative salts of the compounds of Formula (I) include the conventional non-toxic salts and the quaternary ammonium salts which are formed, for example, from inorganic or organic acids or bases by means well known in the art.
  • acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cinnamate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, itaconate, lactate, maleate, mandelate, methane
  • Base salts include alkali metal salts such as potassium and sodium salts, alkaline earth metal salts such as calcium and magnesium salts, and ammonium salts with organic bases such as dicyclohexylamine salts and N-methyl-D-glucamine.
  • basic nitrogen containing groups may be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and strearyl chlorides, bromides, and iodides; aralkyl halides like benzyl and phenethyl bromides and others.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate and diamyl sulfates
  • long chain halides such as decyl, lau
  • the esters in the present invention are non-toxic, pharmaceutically acceptable ester derivatives of the compounds of Formula (I). This may include, for example, ester derivatives prepared from acetic, benzoic, mandelic, stearic, lactic, salicylic, hydroxynaphthoic, glucoheptonic, and gluconic acid.
  • the compounds of Formula (I) may be esterified by a variety of conventional procedures well known by those skilled in the art. For example, esterification may be reached using the appropriate carboxylic acid in the presence of trifluoroacetic anhydride and optionally, pyridine, or in the presence of ⁇ /, ⁇ /-carbonyldiimidazole with pyridine.
  • Sensitive or reactive groups on the compound of Formula (I) may need to be protected during any of the above methods for forming esters, and protecting groups may be added and removed by conventional methods well known in the art.
  • Another object of this invention is to provide methods of making the compounds of the invention.
  • the compounds may be prepared from readily available materials by the methods outlined in the Reaction Schemes and Examples below, and by obvious modifications thereto.
  • compounds of Formula (I), where R is bonded to the pyrimidine ring with a heteroatom such as oxygen or nitrogen may be synthesized by the method illustrated in Reaction Scheme 1.
  • condensation of a ⁇ -ketoester of Formula (II) with a carbamidine of Formula (III) provides the fused pyrimidine azepine of Formula (IV).
  • Treatment of Formula (IV) with a chlorinating agent, such as phosphorus oxychloride provides the pyrimidinyl chloride of Formula (V).
  • R 4 may be labile under the reaction conditions for chlorination, and reinstallation of R 4 may be necessary.
  • R 4 when R 4 is equal to hydrogen, it can be treated with a compound of the Formula R 4 - Ig (where Ig is a leaving group such as iodo, bromo, chloro, or carbonate) in the optional presence of a base to provide the corresponding Formula (V) compound where R 4 is other than hydrogen.
  • Ig is a leaving group such as iodo, bromo, chloro, or carbonate
  • a compound of Formula H-R 2 in the presence of a base such as potassium tert- butoxide or triethylamine, is allowed to react with the chloro compound of Formula (V) in a nucleophilic substitution reaction to provide the compound of Formula (I).
  • a base such as potassium tert- butoxide or triethylamine
  • compounds of Formula (I) where R 2 is bonded to the pyrimidine ring with a carbon atom may also be prepared from the chloride of Formula (V) as outlined in Reaction Scheme I.
  • a compound of Formula (V) can be treated with a compound of Formula BrMg - R 2 in the presence of an iron (III) catalyst, such as iron(lll) acetylacetonate to provide a compound of Formula (I) where R 2 is an alkyl group.
  • an iron (III) catalyst such as iron(lll) acetylacetonate
  • Salts for example, a hydrochloride or a fumaric acid salt
  • a protic acid such as hydrochloric acid or fumaric acid.
  • R 4 of Formula (Ib) [(l)a, where R 4 is hydrogen] may be converted from hydrogen to a group other than hydrogen as shown in Reaction Scheme 2.
  • a compound of Formula (Ib) where R 4 is hydrogen may be treated with a base, such as sodium bicarbonate, to provide a compound of Formula (Ic) where R 4 is hydrogen.
  • the compound of Formula (Ic) where R 4 is hydrogen may then undergo reaction with a compound of Formula R 4 - Ig (where Ig is a leaving group such as iodo, bromo, chloro, or carbonate), in the optional presence of a base, to provide compounds of Formula (Id) where R 4 is other than hydrogen.
  • the compound of Formula (II) where one R 3 is 3-methyl may be prepared from the beta-alanine derivative (Vl).
  • This compound of Formula (VI) can be deprotonated with a base, such as sodium hydride, and allowed to react with an alkylating agent of Formula (VII) to yield a compound of Formula (VIII).
  • the ⁇ , ⁇ -unsaturated ester of Formula (VIII) may then undergo a tandem reduction and cyclization in the presence of an electron transfer source such as magnesium, and in an alcoholic solvent such as methanol to provide the compound of Formula (Ha).
  • the process may be completed in stepwise fashion by treating the compound of Formula (VIII) with a reducing agent, such as sodium borohydride, to give the diester of Formula (IX).
  • a reducing agent such as sodium borohydride
  • the compound of Formula (IX) can then undergo a Dieckmann condensation when treated with a base such as sodium methoxide, to generate the compound of Formula (Ha).
  • the ketoester of Formula (lib), wherein the R 3 at position 3 can be either a hydrogen or a 3-methyl group may be methylated to give the compound of Formula (lie) by quenching the dianion of Formula (lib), formed by treatment of Formula (lib) with strong base, with a methylating agent such as methyl iodide.
  • the dimethyl compound of Formula (lid) is likewise formed by quenching the dianion of (lie) with methyl iodide.
  • HPLC-electrospray mass spectra were obtained using a Hewlett Packard 1100 HPLC equipped with a quaternary pump, a variable wavelength detector, a YMC Pro 18 2.0 mm x 23 mm column, and a Finnigan LCQ ion trap mass spectrometer with electrospray ionization. Gradient elution from 90% A to 95% B over 5 minutes was used on the HPLC. Buffer A was 98% water, 2% Acetonitrile, and 0.02% TFA, and Buffer B was 98% Acetonitrile, 2% water, and 0.018% TFA. Spectra were scanned from 140- 1200 amu using a variable ion time according to the number of ions in the source.
  • PS-DIEA polystyrene-bound diisopropylethylamine
  • Step 1 Preparation of tert-butyl 2-cvclopropyl-4-hvdroxy-5,6,8,9-tetrahydro-7H - pyrimido[4,5-d]azepine-7-carboxylate
  • Step 2 Preparation of tert-butyl 4-chloro-2-cvclopropyl-5,6,8,9-tetrahydro-7H - pyrimidof4,5-cnazepine-7-carboxylate
  • Step 3 Preparation of tert-butyl 2-cvclopropyl-4-(3,3,3-trifluoropropoxy)-5,6,8,9- tetrahydro-7H-pyrimido[4,5-d]azepine-7-carboxylate
  • Step 1 Preparation of 1-tert-butyl 4-ethyl 6-methyl-5-oxoazepane-1 ,4- dicarboxylate
  • lodomethane (240 ⁇ L, 3.85 mmol) was added, and the mixture was allowed to return to rt. An excess of water was added, and the mixture was extracted with ethyl acetate. The organic layer was separated, dried over anhydrous sodium sulfate, and concentrated in vacuo. The resulting crude material was purified using Biotage® flash 4OS (2:1 hexanes / ethyl acetate) to afford 250 mg (24%) of the desired product.
  • Step 2 Preparation of tert-butyl 2-cvclopropyl-4-hvdroxy-9-methyl-5,6,8,9- tetrahvdro-7/-/-pyrimido[4,5-d]azepine-7-carboxylate
  • Step 3 Preparation of 4-chloro-2-cvclopropyl-9-methyl-6,7.8,9-tetrahydro-5H- pyrimido[4,5-d]azepine
  • Step 4 Preparation of tert-butyl 4-chloro-2-cvclopropyl-9-methyl-5,6,8,9- tetrahvdro-7H-pyrimido[4,5-d]azepine-7-carboxylate
  • Step 5 Preparation of 2-cvclopropyl-9-methyl-4-(3.3,3-trifluoropropoxy)-6,7,8,9- tetrahvdro-5H-pyrimido[4,5-d]azepine dihvdrochloride
  • the solution was stirred at ambient temperature for 4 h.
  • the contents of the flask were partitioned between ethyl acetate and water.
  • the organic layer was separated and concentrated to a crude oil under reduced pressure.
  • the resulting crude was purified by HPLC eluted with an acetonitrile / water mixture to provide the title compound as a yellow solid (82 mg, 99%).
  • subject includes mammals (e.g., humans and animals).
  • treatment includes any process, action, application, therapy, or the like, wherein a subject, including a human being, is provided medical aid with the object of improving the subject's condition, directly or indirectly, or slowing the progression of a condition or disorder in the subject.
  • combination therapy means the administration of two or more therapeutic agents to treat a disease, condition, and/or disorder.
  • Such administration encompasses co-administration of two or more therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each inhibitor agent.
  • additioi such administration encompasses use of each type of therapeutic agent in a sequential manner.
  • terapéuticaally effective means the amount of each agent administered that will achieve the goal of improvement in a disease, condition, and/or disorder severity, while avoiding or minimizing adverse side effects associated with the given therapeutic treatment.
  • pharmaceutically acceptable means that the subject item is appropriate for use in a pharmaceutical product.
  • an embodiment of this invention includes a method of treating the various conditions in a patient (including mammals) which comprises administering to said patient a composition containing an amount of the compound of Formula (I) that is effective in treating the target condition.
  • the compounds of the present invention interact with the 5-HT 2C receptor and may be used in the treatment or prevention of diseases and/or behaviors that involve the 5-HT 2C receptor.
  • diseases and/or behaviors include, for example, obesity, obesity related disorders such as diabetes, feeding behavior, eating disorders such as bulimia, anorexia nervosa and premenstrual tension.
  • an object of this invention is to provide methods for treating obesity and inducing weight loss in an individual by administration of a compound of the invention.
  • the method of the invention comprises administering to an individual a therapeutically effective amount of at least one compound of the invention, or a prodrug thereof, which is sufficient to induce weight loss.
  • the invention further comprises a method of preventing weight gain in an individual by administering an amount of at least one compound of the invention, or a prodrug thereof, which is sufficient to prevent weight gain.
  • Further diseases and/or behaviors which can be treated or prevented include central nervous disorders, depressions, anxiety disorders, obsessive-compulsive disorders, sleep disorders, sexual dysfunction, psychoses, migraine, schizophrenia, drug or alcohol addiction and chronic fatigue syndrome.
  • the present invention also relates to the use of the compounds of this invention for the treatment of obesity-related diseases including associated dyslipidemia and other obesity- and overweight-related complications such as, for example, cholesterol gallstones, gallbladder disease, gout, cancer (e.g., colon, rectum, prostate, breast, ovary, endometrium, cervix, gallbladder, and bile duct), menstrual abnormalities, infertility, polycystic ovaries, osteoarthritis, and sleep apnea, as well as for a number of other pharmaceutical uses associated therewith, such as the regulation of appetite and food intake, dyslipidemia, hypertriglyceridemia, Syndrome X, type 2 diabetes (non-insulin- dependent diabetes), atherosclerotic diseases such as heart failure, hyperlipidemia, hypercholesteremia, low HDL levels, hypertension, cardiovascular disease (including atherosclerosis, coronary heart disease, coronary artery disease, and hypertension), cerebrovascular disease such as stroke,
  • Compounds of Formula (I) may be administered alone or in combination with one or more additional therapeutic agents.
  • Combination therapy includes administration of a single pharmaceutical dosage formulation which contains a compound of Formula (I) and one or more additional therapeutic agents, as well as administration of the compound of Formula (I) and each additional therapeutic agents in its own separate pharmaceutical dosage formulation.
  • a compound of Formula (I) and a therapeutic agent may be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent may be administered in separate oral dosage formulations.
  • the compound of Formula (I) and one or more additional therapeutic agents may be administered at essentially the same time (e.g., concurrently) or at separately staggered times (e.g., sequentially).
  • anti-obesity drugs include ⁇ -3 adrenergic receptor agonists such as CL 316,243; cannabinoid (e.g., CB-1) antagonists such as Rimonabant; neuropeptide-Y receptor antagonists; neuropeptide Y5 inhibitors; apo-B/MTP inhibitors; 11 ⁇ -hydroxy steroid dehydrogenase-1 inhibitors; peptide YY 3 - 36 or analogs thereof; MCR4 agonists; CCK-A agonists; monoamine reuptake inhibitors; sympathomimetic agents; dopamine agonists; melanocyte-stimulating hormone receptor analogs; melanin concentrating hormone antagonists; leptin; leptin analogs; leptin receptor agonists; galanin antagonists; lipase inhibitors; bombesin agonists; thyromim
  • the compound of Formula (I) may be administered in combination with one or more of the following agents for the treatment of diabetes or diabetes-related disorders including PPAR ligands (agonists, antagonists), insulin secretagogues, for example, sulfonylurea drugs and non-sulfonylurea secretagogues, ⁇ -glucosidase inhibitors, insulin sensitizers, hepatic glucose output lowering compounds, and insulin and insulin derivatives.
  • Such therapies may be administered prior to, concurrently with, or following administration of the compounds of the invention.
  • Insulin and insulin derivatives include both long and short acting forms and formulations of insulin.
  • PPAR ligands may include agonists and/or antagonists of any of the PPAR receptors or combinations thereof.
  • PPAR ligands may include ligands of PPAR- ⁇ , PPAR- ⁇ , PPAR- ⁇ or any combination of two or three of the receptors of PPAR.
  • PPAR ligands include, for example, rosiglitazone, troglitazone, and pioglitazone.
  • Sulfonylurea drugs include, for example, glyburide, glimepiride, chlorpropamide, tolbutamide, and glipizide, ⁇ - glucosidase inhibitors that may be useful in treating diabetes when administered with a compound of the invention include acarbose, miglitol, and voglibose.
  • PPAR- ⁇ agonists such as the glitazones (e.g., troglitazone, pioglitazone, englitazone, MCC-555, rosiglitazone, and the like) and other thiazolidinedione and non-thiazolidinedione compounds
  • biguanides such as metform
  • Hepatic glucose output lowering compounds that may be useful in treating diabetes when administered with a compound of the invention include glucagon anatgonists and metformin, such as Glucophage and Glucophage XR.
  • Insulin secretagogues that may be useful in treating diabetes when administered with a compound of the invention include sulfonylurea and non-sulfonylurea drugs: GLP-1 , GIP, PACAP, secretin, and derivatives thereof; nateglinide, meglitinide, repaglinide, glibenclamide, glimepiride, chlorpropamide, glipizide.
  • GLP-1 includes derivatives of GLP-1 with longer half-lives than native GLP-1 , such as, for example, fatty-acid derivatized GLP-1 and exendin.
  • Compounds of the invention may also be used in methods of the invention in combination with drugs commonly used to treat lipid disorders in patients.
  • drugs include, but are not limited to, HMG-CoA reductase inhibitors, nicotinic acid, fatty acid lowering compounds (e.g., acipimox); lipid lowering drugs (e.g., stanol esters, sterol glycosides such as tiqueside, and azetidinones such as ezetimibe), ACAT inhibitors (such as avasimibe), bile acid sequestrants, bile acid reuptake inhibitors, microsomal triglyceride transport inhibitors, and fibric acid derivatives.
  • HMG-CoA reductase inhibitors e.g., nicotinic acid, fatty acid lowering compounds (e.g., acipimox); lipid lowering drugs (e.g., stanol esters, sterol glycosides such as
  • HMG-CoA reductase inhibitors include, for example, lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rivastatin, itavastatin, cerivastatin, and ZD-4522.
  • Fibric acid derivatives include, for example, clofibrate, fenofibrate. bezafibrate, ciprofibrate, beclofibrate, etofibrate, and gemfibrozil.
  • Sequestrants include, for example, cholestyramine, colestipol, and dialkylaminoalkyl derivatives of a cross-linked dextran.
  • Compounds of the invention may also be used in combination with anti ⁇ hypertensive drugs, such as, for example, ⁇ -blockers and ACE inhibitors.
  • additional anti-hypertensive agents for use in combination with the compounds of the present invention include calcium channel blockers (L-type and T-type; e.g., diltiazem, verapamil, nifedipine, amlodipine and mybefradil), diuretics (e.g., chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzthiazide, ethacrynic acid tricrynafen, chlorthalidone, furosemide, musolimine, bumetanide, triamtrenene, amiloride, spironolactone),
  • compositions which are comprised of an inert carrier and an effective amount of a compound of Formula (I), or a salt, or ester thereof.
  • An inert carrier is any material which does not interact with the compound to be carried and which lends support, means of conveyance, bulk, traceable material, and the like to the compound to be carried.
  • An effective amount of the compound is that amount which produces a result or exerts an influence on the particular procedure being performed.
  • prodrug forms of the compounds of this invention will prove useful in certain circumstances, and such compounds are also intended to fall within the scope of the invention.
  • Prodrug forms may have advantages over the parent compounds exemplified herein, in that they are better absorbed, better distributed, more readily penetrate the central nervous system, are more slowly metabolized or cleared, etc.
  • Prodrug forms may also have formulation advantages in terms of crystallinity or water solubility.
  • compounds of the invention having one or more hydroxyl groups may be converted to esters or carbonates bearing one or more carboxyl, hydroxyl or amino groups, which are hydrolyzed at physiological pH values or are cleaved by endogenous esterases or lipases in vivo (see, e.g., U.S. Patent Nos. 4,942,184; 4,960,790; 5,817,840; and 5,824,701 , all of which are incorporated herein by reference in their entirety, and references therein).
  • the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication.
  • the amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
  • the total amount of the active ingredient to be administered may generally range from about 0.001 mg/kg to about 200 mg/kg.
  • a unit dosage may contain from about 0.05 mg to about 1500 mg of active ingredient, and may be administered one or more times per day.
  • the daily dosage for administration by injection including intravenous, intramuscular, subcutaneous, and parenteral injections, and use of infusion techniques may be from about 0.01 to about 200 mg/kg.
  • the daily rectal dosage regimen may be from 0.01 to 200 mg/kg of total body weight.
  • the transdermal concentration may be that required to maintain a daily dose of from 0.01 to 200 mg/kg.
  • the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age of the patient, the diet of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like.
  • the desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt thereof may be ascertained by those skilled in the art using conventional treatment tests.
  • the compounds of this invention may be utilized to achieve the desired pharmacological effect by administration to a subject in need thereof in an appropriately formulated pharmaceutical composition.
  • a subject for example, may be a mammal, including a human, in need of treatment for a particular condition or disease. Therefore, the present invention includes pharmaceutical compositions which are comprised of a pharmaceutically acceptable carrier and a pharmaceutically effective amount of a compound identified by the methods described herein, or a pharmaceutically acceptable salt or ester thereof.
  • a pharmaceutically acceptable carrier is any carrier which is relatively non-toxic and innocuous to a patient at concentrations consistent with effective activity of the active ingredient so that any side effects ascribable to the carrier do not vitiate the beneficial effects of the active ingredient.
  • a pharmaceutically effective amount of a compound is that amount which produces a result or exerts an influence on the particular condition being treated.
  • the compounds identified by the methods described herein may be administered with a pharmaceutically-acceptable carrier using any effective conventional dosage unit forms, including, for example, immediate and timed release preparations, orally, parenterally, topically, or the like.
  • the compounds may be formulated into solid or liquid preparations such as, for example, capsules, pills, tablets, troches, lozenges, melts, powders, solutions, suspensions, or emulsions, and may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions.
  • the solid unit dosage forms may be a capsule which can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and corn starch.
  • the compounds of this invention may be tableted with conventional tablet bases such as lactose, sucrose, and cornstarch in combination with binders such as acacia, cornstarch, or gelatin; disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum; lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example, talc, stearic acid, or magnesium, calcium or zinc stearate; dyes; coloring agents; and flavoring agents intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient.
  • conventional tablet bases such as lactose, sucrose, and cornstarch in combination with binders such as acacia, cornstarch, or gelatin
  • disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and
  • Suitable excipients for use in oral liquid dosage forms include diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent.
  • diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent.
  • Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both.
  • Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example, those sweetening, flavoring and coloring agents described above, may also be present.
  • the pharmaceutical compositions of this invention may also be in the form of oil- in-water emulsions.
  • the oily phase may be a vegetable oil such as liquid paraffin or a mixture of vegetable oils.
  • Suitable emulsifying agents may be naturally occurring gums, naturally occurring phosphatides, esters or partial esters derived from fatty acids and hexitol anhydrides, and condensation products of said partial esters.
  • the emulsions may also contain sweetening and flavoring agents.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil such or in a mineral oil.
  • the oily suspensions may contain a thickening agent.
  • the suspensions may also contain one or more preservatives, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents.
  • Syrups and elixirs may be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol, or sucrose. Such formulations may also contain a demulcent, and preservative, flavoring and coloring agents.
  • sweetening agents such as, for example, glycerol, propylene glycol, sorbitol, or sucrose.
  • Such formulations may also contain a demulcent, and preservative, flavoring and coloring agents.
  • the compounds of this invention may also be administered parenterally, that is, subcutaneously, intravenously, intramuscularly, or interperitoneally, as injectable dosages of the compound in a physiologically acceptable diluent with a pharmaceutical carrier, with or without the addition of a pharmaceutically acceptable surfactants, suspending agents, emulsifying agents or other pharmaceutical adjuvants.
  • compositions of this invention may typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimize or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulation ranges from about 5% to about 15% by weight.
  • the surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB.
  • compositions may be in the form of sterile injectable aqueous suspensions.
  • Such suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent.
  • Diluents and solvents that may be employed are, for example, water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile fixed oils are conventionally employed as solvents or suspending media.
  • any bland, fixed oil may be employed including synthetic mono or diglycerides.
  • fatty acids such as oleic acid may be used in the preparation of injectables.
  • a composition of the invention may also be administered in the form of suppositories for rectal administration of the drug.
  • These compositions may be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such material are, for example, cocoa butter and polyethylene glycol.
  • transdermal delivery devices Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts.
  • transdermal patches for the delivery of pharmaceutical agents is well known in the art (see, e.g., U.S. Patent No. 5,023,252, incorporated herein by reference).
  • patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • compositions of the invention may also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired. Any of the compositions of this invention may be preserved by the addition of an antioxidant such as ascorbic acid or by other suitable preservatives. Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized.
  • the compounds identified by the methods described herein may be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutical agents where the combination causes no unacceptable adverse effects.
  • the compounds of this invention can be combined with known anti-obesity, or with known antidiabetic or other indication agents, and the like, as well as with admixtures and combinations thereof.
  • compositions which are comprised of an inert carrier and an effective amount of a compound identified by the methods described herein, or a salt or ester thereof.
  • An inert carrier is any material which does not interact with the compound to be carried and which lends support, means of conveyance, bulk, traceable material, and the like to the compound to be carried.
  • An effective amount of compound is that amount which produces a result or exerts an influence on the particular procedure being performed.
  • Formulations suitable for subcutaneous, intravenous, intramuscular, and the like; suitable pharmaceutical carriers; and techniques for formulation and administration may be prepared by any of the methods well known in the art (see, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 20 th edition, 2000)
  • Demonstration of the activity of the compounds of the present invention may be accomplished through in vitro, ex vivo, and in vivo assays that are well known in the art. For example, to demonstrate the activity of the compounds of the present invention, the following assays may be used.
  • Activity of the compounds of Formula (I) may be determined by measurement of their binding affinity and agonist activity at the 5-HT 2C receptor. The following assays were performed to determine the effect of the compounds of Formula (I) on the 5-HT 2C receptor.
  • the functional activity of compounds of Formula (I) was measured using a FLIPR calcium mobilization assay in cells transfected with recombinant receptor.
  • AV-12 cells expressing 5-HT 2C receptors were placed into 96-well black plates (5 x10 4 cells per well/ 100 ⁇ L DMEM medium.
  • the DMEM medium contained 10% dialysis FBS, 2 mM L- glutamine, 20 mM Hepes, 1 mg/ml G418, and 1 % antibiotic.
  • the cell plates are incubated overnight at 37°C, 5% of CO 2 .
  • the cell culture medium was removed, and 160 ⁇ L of 1X loading buffer (1X Hank's buffer contains 20 mM Hepes and 2.5 mM probenecid dissolved in 0.5 N NaOH) was added with calcium dye (Calcium 3 Assay Kit, Molecular Devices, Inc) into the cell plates.
  • the cells were incubated at 37°C, 5% CO 2 for 1 hour. Thereafter, the cell plates and compound dilution plates were placed into the FLIPR instrument, and 40 ⁇ L of varying concentrations of test compound or reference compound (alpha-Me-5-HT) were added into the cell plates. Increases in intracellular calcium were recorded at 488 nm during 3 minutes with the FLIPR.
  • Peak fluorescence counts minus basal counts from 1 sec to 180 sec time points were used to determine agonist activity. Iterative curve-fitting and parameter estimations were analyzed using a four-parameter logistic equation and a program that generates dose- response curves. The potency of the compound was expressed in terms of the concentration required to achieve half-maximal stimulation (EC 50 ) while the efficacy of the ligand was expressed as the percentage of its maximal stimulation relative to the maximal stimulation observed with the alpha-Me-5-HT curve from the same plate (E/Emax).
  • the purpose of this protocol is to determine the effect of a single dose of an unknown compound on food consumption of lean overnight fasted rats.
  • the fasted-refed rat model is frequently used in the field of obesity to identify compounds with potential for anorectic effects.
  • This animal model has been successfully used in the identification and characterization of the efficacy profile of compounds that are or have been used in the management of body weight in obese humans (see, e.g., Balvet, et al., Gen. Pharmacol. 13:293-297, 1982; ⁇ rignaschi, et al., Br. J. Pharmacol. 127:1190-1194, 1999; McTavish and Heel, Drug 43:713-733, 1992; Rowland, et al., Life Sci. 36:2295-2300, 1985).
  • the rats are fasted overnight during the dark phase (total of approx. 16-18 hr).
  • the animal is dosed with an assigned treatment (2 mg/mL).
  • One hour after dosing pre- weighed food jars are returned to the cage. Food intake is recorded 30, 60, 90, 180, and 240 minutes and 24 hours post-food return.
  • spillage is returned to the food jar and then the food jars are weighed. The amount of food consumed is determined for each time point. Difference between treatment group is determined using appropriate statistical analysis.
  • the purpose of this protocol is to determine the effect of chronic administration of an unknown compound on body weight and food and water consumption in obese Zucker (fa/fa) rats.
  • Obese Zucker rats are frequently used in the determination of compound efficacy in the reduction of body weight.
  • This animal model has been successfully used in the identification and characterization of the efficacy profile of compounds that are or have been used in the management of body weight in obese humans (see, e.g., Al- Barazanji, et al., Obes Res. 8:317-323, 2000; Assimacopoulos-Jeannet, et al., Am. J. Physiol. 260(2 Pt 2):R278-283, 1991 ; Dryden, et al., Horm.
  • a typical study includes 60-80 male Zucker (fa/fa) rats (approximatelylO/treatment group) with an average body weight of approximately 550 g. Rats are kept in standard animal rooms under controlled temperature and humidity and a 12/12 light/dark cycle. Water and food are continuously available. Rats are single-housed in large rat shoeboxes containing grid floor. Animals are adapted to the grid floors and sham-dosed with study vehicle for at least four days before the recording of two-days baseline measurement of body weight and 24 h food and water consumption. Rats are assigned to one of 6-8 treatment groups based upon their baseline body weight. The groups are set up so that the mean and standard error of the mean of body weight are similar.
  • Animals are orally gavaged (2 ml_/kg) daily before the dark phase of the light/dark cycle for a pre-determined number of days (typically 6-14 days) with their assigned dose/compound. At this time, body weight, food and water consumption are measured. On the final day, body weight is measured and animals are euthanized by CO 2 inhalation.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to tetrahydro-5H-pyrimido[4,5-d]azepine compounds of Formula (I) that are useful in the treatment or prevention of diseases and/or behaviors that involve the 5-HT2C receptor. The compounds of the present invention may also be used for the treatment or prevention of diseases and/or disorders including obesity, obesity-related disorders such as diabetes, feeding behavior disorders, eating disorders such as bulimia and anorexia nervosa, and premenstrual tension.

Description

TETRAHYDRO-5H-PYRIMIDO[4,5-d]AZEPINE DERIVATIVES USEFUL FOR THE TREATMENT OF DISEASES ASSOCIATED WITH THE 5-HT2C RECEPTOR
FIELD OF THE INVENTION
[001] This application claims benefit of U.S. Provisional Application Serial No. 60/619,195; filed on October 15, 2004, the contents of which are incorporated herein by reference in their entirety.
[002] The invention relates to tetrahydro-5H -pyrimido[4,5-d]azepine compounds of Formula (I) that are useful in the treatment or prevention of diseases and/or behaviors that involve the 5-HT2C receptor. The compounds of the present invention may also be used for the treatment or prevention of diseases and/or disorders including obesity, obesity-related disorders such as diabetes, feeding behavior disorders, eating disorders such as bulimia and anorexia nervosa, and premenstrual tension.
BACKGROUND OF THE INVENTION
[003] Obesity, which is an excess of body fat relative to lean body mass, is a chronic disease that is highly prevalent in modem society. It is associated not only with a social stigma, but also with decreased life span and numerous medical problems, including adverse psychological development, coronary artery disease, hypertension, stroke, diabetes, hyperlipidaemia, and some cancers (see, e.g., Nishina, et al., Metab. 43:554- 558, 1994; Grundy and Barnett, Dis. Mon. 36:641-731 , 1990; Rissanen, et al., British Medical Journal, 301 :835-837, 1990).
[004] Obesity is considered a major medical problem largely because it is a factor for a number of other diseases. For example, obesity is correlated with a much higher incidence of type 2 diabetes (non-insulin dependent diabetes mellitus, NIDDM), hypertension, hyperlipidemia, myocardial infarction, cancers, gallbladder disease, respiratory disease, gout, arthritis, and dermatological disease. In addition, obese individuals have a higher incidence of mortality at a younger age as compared to their leaner counterparts.
[005] Serotonin (5-HT) has been implicated in the regulation of feeding behavior. Drugs which increase the concentration of 5-HT in the synaptic cleft by increasing 5-HT release and/or inhibiting re-uptake of the transmitter (such as Redux® (dexfenfluramine) and sibutramine) are effective long-term treatments for obesity, albeit with significant side-effects.
[006] Serotonin produces physiological effects by acting on a heterogeneous family of receptors. The lack of selective agonists and antagonists for the individual subtypes of serotonin receptors has prevented a complete characterization of the physiological role of each receptor subtype.
[007] Regulation of several subtypes of 5-HT receptors (5-HT1A, 5-HT1B, 5-HT2A, 5-HT2B, 5-HT6, and 5-HT2C) has been demonstrated to elicit effects on food intake. For example, activation of both 5-HT2A and 5-HT2C receptors decrease food intake. However, while 5- HT2A receptor agonists are thought to decrease food intake by disrupting the ability of the animal to feed, the 5-HT2C receptor has been implicated in the regulation of satiety. 5- HT2C receptors are localized to the hypothalamus and the brainstem, two brain regions known to play a critical role in the modulation of food intake. Furthermore, data available to date suggests that 5-HT2C receptor agonists produce a decrease in food intake which is associated with the least likely potential for side effects.
[008] Targeting the 5-HT2C receptor for the treatment of obesity and feeding disorders has previously been described (Kennett & Curzon, Psychopharmacology 96(1):93-100, 1988; Curzon, Adv. Exp. Med. Biol. 294:377-88, 1991 ; Walsh, et al., Psychopharmacology 16(1 ):120-2, 1994; Dourish, Obes. Res. Suppl. 4:449-462, 1995; Sargent, et al., Psychopharmacology 133:309-312,1997). It would be expected that agonists that are selective for this receptor would have superior properties with respect to other known appetite suppressants, such as serotonin/noradrenaline re-uptake inhibitors and/or releasers, which can lead to hypertension and/or cardiac valve defects.
[009] Non-selective agonists/partial agonists (e.g., mCPP, TFMPP) of the 5-HT2C receptor have been shown to reduce food intake in rats and to accelerate the appearance of the behavioral satiety sequence. Importantly, the hypophagic effects of mCPP are antagonized by the selective 5-HT2C receptor antagonist, SB-242084. Recent findings from studies in normal human volunteers and obese subjects administered mCPP have also shown decreases in food intake. A single injection of mCPP decreased food intake in female volunteers and subchronic treatment for a 14 day period decreased the appetite and body weight of obese male and female subjects.
[010] The importance of the 5-HT2C receptor in mediating feeding behavior is further supported by studies on mutant 5-HT2C-knockout mice (Tecott, et al., Nature 374:542- 546, 1995; Heath & Hen, Curr. Biol. 5(9):997-999, 1995). Interestingly, the knockout mice show significantly greater weight gain and adipose tissue deposits over time compared to wild-type mice. Additional studies have confirmed that 5-HT2C knockout mice overeat and become obese which appears to be due to a defect in their satiety mechanism. In the behavioral satiety sequence model, knockout animals continued to eat for a significantly longer period of time than the wild-type controls. The prolonged eating in the 5-HT2C receptor knockout mice was enhanced by access to a sweet diet, suggesting that the 5- HT2C receptor may also play a role in palatability.
[011] It is significant that the decrease in food intake induced by dexfenfluramine is markedly attenuated in 5-HT2C receptor knockout mice. These results suggest that dexfenfluramine enhances satiety and decreases food intake via an agonist action on 5- HT2C receptors. In addition, in wild-type animals, these anorectic effects of dexfenfluramine are blocked by the 5-HT2C-selective antagonist, SB-242084. These data are consistent with the clinical evidence that the anorectic effect of dexfenfluramine was blocked by the 5HT2 receptor antagonist, ritanserin.
[012] Thus, animal and human data strongly implicate the involvement of the 5-HT2C receptor in satiety, and compounds that interact with this receptor would be effective in treating diseases and/or disorders such as obesity, eating disorders, and the like.
[013] The compounds of the present invention interact with the 5-HT2C receptor. Thus, these compounds may be useful for treatment or prevention of diseases and/or behaviors associated with the 5-HT2C receptor. Furthermore, the compounds of the present invention may also be used for the treatment and/or prevention of obesity, obesity-related disorders such as diabetes, feeding behavior, eating disorders such as bulimia and anorexia nervosa, and premenstrual tension. In addition, other diseases and/or disorders such as central nervous disorders, depression, anxiety disorders, obsessive-compulsive disorders, sleep disorders, sexual dysfunction, psychoses, migraine, schizophrenia, drug or alcohol addiction, and chronic fatigue syndrome may also be treated and/or prevented by the compounds of the present invention.
DETAILED DESCRIPTION OF THE INVENTION
[014] The invention relates to a compound of Formula (I)
Figure imgf000004_0001
wherein R1 is
• (C1-C4)alkyl,
• phenyl optionally substituted with halo, or
• (C3-C6) cycloalky I optionally substituted with (C1-C3)alkyl;
R2 is
• C1-C6 alkyl,
• halo
• piperidin-1-yl
• pyrrolidin-1-yl
• phenylpyrrolidin-1 -yl optionally substituted on the phenyl ring with up to two groups selected from halo, (C1-C3)alkyl, (C1-C3)alkoxy, or CF3,
• phenylpiperidin-1-yl optionally substituted on the phenyl ring with up to two groups selected fromhalo or CF3,
• phenoxypiperidin-1 -yl optionally substituted on the phenyl ring with up to two groups selected from halo or CF3,
• phenylpiperazin-1 -yl optionally substituted on the phenyl ring with up to two groups selected from (C1-C3)alkyl, halo, CF3, or (C1-C3)alkoxy,
• -NR5R6 wherein
R5 is H or (C1-C3)alkyl, and
R6 is (C1-C3)alkyl optionally substituted with
• indoly-2-yl optionally substituted with halo,
• phenyl optionally substituted with up to two groups selected from (C1-C3)alkoxy, or benzodioxan-6-yl, or
• OR7 wherein
R7 is
• H1
• (C3-C6)cycloalkyl optionally substituted with (C1-C3)alkyl,
• 2,3-dihydro-inden-2-yl,
• tetrahydrofuran-yl, or
• (C1-C6)alkyl optionally substituted with
• F up to the perfluoro level,
• (C3-C6)cycloalkyl,
• (C1-C3)alkoxy,
• — N[(C1-C3)alkyl]2, . — NH[(C1-C3)alkyl],
• morpholin-1-yl,
• phenyl optionally substituted with up to two groups selected from CF3, halo, (C1-C3)alkyl, NO2, (C1-C3)alkoxy, or N[(C1-C3)alkyl]2,
• 2,3-dihydro-benzo[1 ,4]dioxin-2-yl,
• 2,3-dihydro-1 H-indol-2-yl,
• tetrahydrofuran-yl,
• 2-oxo-2,3-dihydro-benzoimidazol-1 -yl,
• 2-oxo-pyrrolidin-1-yl, or
• — N[(C1-C3)alkyl]phenyl, wherein the phenyl is optionally substituted with up to two groups selected from CF3, halo, (C1-C3)alkyl, (C1- C3)alkoxy, and — N[(C1-C3)alkyl]2;
R3 is H or (C1-C3)alkyl; and
R4 is H, methyl, -C(O)O-R8, or -C(O)R9; wherein
R8 is (C1-C4)alkyl or phenyl optionally substituted with halo, (C1-C3)alkyl, (C1-C3)alkoxy, or CF3, and
R9 is (C1-C4)alkyl or phenyl optionally substituted with halo, (C1-C3)alkyl, (C1-C3)alkoxy, or CF3;
or pharmaceutically acceptable salts thereof.
Definitions
[015] The terms identified above have the following meaning throughout:
[016] The terms "(C1-C3)alkyl," "(C1-C4)alkyl," and (C1-C6)alkyl" mean a linear or branched saturated hydrocarbon groups having from 1 to about 3, from 1 to about 4, and from 1 to about 6 carbon atoms, respectively. Such groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, pentyl, hexyl, and the like.
[017] The term "(C1-C3)alkoxy" means a linear or branched saturated hydrocarbon group having from about 1 to about 3 carbon atoms, said group being attached to an oxygen atom. The oxygen atom is the atom through which the alkoxy substituent is attached to the rest of the molecule. Such groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, and the like.
[018] The term "halo" means F, Br, Cl, and I.
[019] The term "(C3-C6)cycloalkyl" means a saturated monocyclic alkyl group of from 3 to about 6 carbon atoms and includes, but is not limited to, such groups as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
[020] The term "optionally substituted" means that the moiety so modified may be unsubstituted or substituted with the identified substituent(s). When any moiety is described as being substituted, it can have one or more of the indicated substituents that may be located at any available position on the moiety. When there are two or more substituents on any moiety, each substituent may be defined independently of any other substituent and may, accordingly, be the same or different.
[021] Each substituent may replace any H atom on the moiety so modified as long as the replacement is chemically possible and chemically stable. When there are two or more substituents on any moiety, each substituent is chosen independently of any other substituent and can, accordingly, be the same or different.
[022] The compounds of Formula (I) may contain one or more asymmetric centers, depending upon the location and nature of the various substituents desired. Asymmetric carbon atoms may be present in the (R) or (S) configuration. Preferred isomers are those with the absolute configuration which produces the compound of Formula (I) with the more desirable biological activity. In certain instances, asymmetry may also be present due to restricted rotation about a given bond, for example, the central bond adjoining two aromatic rings of the specified compounds.
[023] Substituents on a ring may also be present in either cis or trans form, and a substituent on a double bond may be present in either Z or E form.
[024] It is intended that all isomers (including enantiomers arid diastereomers), either by nature of asymmetric centers or by restricted rotation as described above, as separated, pure or partially purified isomers or racemic mixtures thereof, be included within the scope of the instant invention. The purification of said isomers and the separation of said isomeric mixtures may be accomplished by standard techniques known in the art.
[025] Representative salts of the compounds of Formula (I) include the conventional non-toxic salts and the quaternary ammonium salts which are formed, for example, from inorganic or organic acids or bases by means well known in the art. For example, such acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cinnamate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, itaconate, lactate, maleate, mandelate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, pamoate, pectinate persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, sulfonate, tartrate, thiocyanate, tosylate, and undecanoate.
[026] Base salts include alkali metal salts such as potassium and sodium salts, alkaline earth metal salts such as calcium and magnesium salts, and ammonium salts with organic bases such as dicyclohexylamine salts and N-methyl-D-glucamine. Additionally, basic nitrogen containing groups may be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and strearyl chlorides, bromides, and iodides; aralkyl halides like benzyl and phenethyl bromides and others.
[027] The esters in the present invention are non-toxic, pharmaceutically acceptable ester derivatives of the compounds of Formula (I). This may include, for example, ester derivatives prepared from acetic, benzoic, mandelic, stearic, lactic, salicylic, hydroxynaphthoic, glucoheptonic, and gluconic acid. The compounds of Formula (I) may be esterified by a variety of conventional procedures well known by those skilled in the art. For example, esterification may be reached using the appropriate carboxylic acid in the presence of trifluoroacetic anhydride and optionally, pyridine, or in the presence of Λ/,Λ/-carbonyldiimidazole with pyridine. One skilled in the art would readily know how to successfully carry out these as well as other methods of esterification. Sensitive or reactive groups on the compound of Formula (I) may need to be protected during any of the above methods for forming esters, and protecting groups may be added and removed by conventional methods well known in the art.
[028] Another object of this invention is to provide methods of making the compounds of the invention. The compounds may be prepared from readily available materials by the methods outlined in the Reaction Schemes and Examples below, and by obvious modifications thereto.
[029] The particular method to be utilized in the preparation of the compounds of this invention depends upon the specific compound desired. Such factors as the selection of the specific moieties and the specific substituents on the various moieties, all play a role in the path to be followed in the preparation of the specific compounds of this invention. These factors are readily recognized by one of ordinary skill in the art.
[030] For synthesis of any particular compound, one skilled in the art will recognize that the use of protecting groups may be required for the synthesis of compounds containing certain substituents. A description of suitable protecting groups and appropriate methods of adding and removing such groups may be found in the art (see, e.g., T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis; Wiley: New York, 1999).
[031] In the Reaction Schemes below, one skilled in the art will recognize that reagents and solvents actually used may be selected from several reagents and solvents well known in the art to be effective equivalents. When specific reagents or solvents are shown in a reaction scheme, therefore, they are meant to be illustrative examples of conditions desirable for the execution of that particular reaction scheme. Abbreviations not identified in accompanying text are listed later in this disclosure under "Abbreviations and Acronyms."
General Methods of Preparation
[032] Compounds of Formula (I) may be prepared by standard means or methods analogous thereto, from starting materials which are either commercially available or prepared by standard means obvious to those skilled in the art. Unless otherwise specified, R1-R4 have the same meanings as described above for Formula (I).
[033] Reaction Scheme 1
Figure imgf000010_0001
[034] For example, compounds of Formula (I), where R is bonded to the pyrimidine ring with a heteroatom such as oxygen or nitrogen, may be synthesized by the method illustrated in Reaction Scheme 1. In this scheme, condensation of a β-ketoester of Formula (II) with a carbamidine of Formula (III) provides the fused pyrimidine azepine of Formula (IV). Treatment of Formula (IV) with a chlorinating agent, such as phosphorus oxychloride, provides the pyrimidinyl chloride of Formula (V). R4 may be labile under the reaction conditions for chlorination, and reinstallation of R4 may be necessary. That is, in Formula (V), when R4 is equal to hydrogen, it can be treated with a compound of the Formula R4 - Ig (where Ig is a leaving group such as iodo, bromo, chloro, or carbonate) in the optional presence of a base to provide the corresponding Formula (V) compound where R4 is other than hydrogen.
[035] A compound of Formula H-R2, in the presence of a base such as potassium tert- butoxide or triethylamine, is allowed to react with the chloro compound of Formula (V) in a nucleophilic substitution reaction to provide the compound of Formula (I).
[036] In addition, compounds of Formula (I) where R2 is bonded to the pyrimidine ring with a carbon atom may also be prepared from the chloride of Formula (V) as outlined in Reaction Scheme I. A compound of Formula (V) can be treated with a compound of Formula BrMg - R2 in the presence of an iron (III) catalyst, such as iron(lll) acetylacetonate to provide a compound of Formula (I) where R2 is an alkyl group.
[037] Salts (for example, a hydrochloride or a fumaric acid salt) of the compounds of Formula (I) may be prepared by stirring compounds of Formula (I) in an ethereal solution with a protic acid such as hydrochloric acid or fumaric acid.
[038] Alternatively, R4 of Formula (Ib) [(l)a, where R4 is hydrogen] may be converted from hydrogen to a group other than hydrogen as shown in Reaction Scheme 2. A compound of Formula (Ib) where R4 is hydrogen may be treated with a base, such as sodium bicarbonate, to provide a compound of Formula (Ic) where R4 is hydrogen. The compound of Formula (Ic) where R4 is hydrogen may then undergo reaction with a compound of Formula R4 - Ig (where Ig is a leaving group such as iodo, bromo, chloro, or carbonate), in the optional presence of a base, to provide compounds of Formula (Id) where R4 is other than hydrogen.
[039] Reaction Scheme 2
Figure imgf000012_0001
[040] Compounds of Formula (II) are either commercially available or are readily available by the synthetic sequences described in Reaction Scheme 3 and Reaction Scheme 4.
[041] Reaction Scheme 3
Figure imgf000012_0002
[042] For example, the compound of Formula (II) where one R3 is 3-methyl may be prepared from the beta-alanine derivative (Vl). This compound of Formula (VI) can be deprotonated with a base, such as sodium hydride, and allowed to react with an alkylating agent of Formula (VII) to yield a compound of Formula (VIII). The α,β-unsaturated ester of Formula (VIII) may then undergo a tandem reduction and cyclization in the presence of an electron transfer source such as magnesium, and in an alcoholic solvent such as methanol to provide the compound of Formula (Ha). Alternatively, the process may be completed in stepwise fashion by treating the compound of Formula (VIII) with a reducing agent, such as sodium borohydride, to give the diester of Formula (IX). The compound of Formula (IX) can then undergo a Dieckmann condensation when treated with a base such as sodium methoxide, to generate the compound of Formula (Ha).
[043] Compounds of Formula (II) in which one R3 group is 6-methyl or two R3 groups are 6,6-dimethyl can be prepared as shown in Reaction Scheme 4.
[044] Reaction Scheme 4
Figure imgf000013_0001
[045] example, the ketoester of Formula (lib), wherein the R3 at position 3 can be either a hydrogen or a 3-methyl group, may be methylated to give the compound of Formula (lie) by quenching the dianion of Formula (lib), formed by treatment of Formula (lib) with strong base, with a methylating agent such as methyl iodide. The dimethyl compound of Formula (lid) is likewise formed by quenching the dianion of (lie) with methyl iodide.
[046] By using these above described methods, the compounds of the invention may be prepared. The following specific examples are presented to further illustrate the invention described herein, but they should not be construed as limiting the scope of the invention in any way. EXPERIMENTAL EXAMPLES OF THE INVENTION General Experimental Procedures
[047] HPLC-electrospray mass spectra (HPLC ES-MS) were obtained using a Hewlett Packard 1100 HPLC equipped with a quaternary pump, a variable wavelength detector, a YMC Pro 18 2.0 mm x 23 mm column, and a Finnigan LCQ ion trap mass spectrometer with electrospray ionization. Gradient elution from 90% A to 95% B over 5 minutes was used on the HPLC. Buffer A was 98% water, 2% Acetonitrile, and 0.02% TFA, and Buffer B was 98% Acetonitrile, 2% water, and 0.018% TFA. Spectra were scanned from 140- 1200 amu using a variable ion time according to the number of ions in the source.
[048] Proton (1H) nuclear magnetic resonance (NMR) spectra were measured with a General Electric G N-Omega 300 (300 MHz) spectrometer with either Me4Si (δ 0.00) or residual protonated solvent (CHCI3 δ 7.26; MeOH δ 3.30; DMSO δ 2.49) as standard.
Abbreviations and Acronyms
[049] When the following are used throughout the disclosure, they have the following meaning:
Biotage® silica gel chromatographic system, Biotage Inc.
BOC tert-butoxycarbonyl n-BuLi n-butyllithium calcd calculated
Cbz carbobenzyloxy
CDI carbonyl diimidazole
CD3OD methanol-d4
Celite® diatomaceous earth filter agent, Celite Corp. cone concentrated d doublet
DMF N,N-dimethy[formamide
DMSO dimethylsulfoxide
DMSO-d6 dimethylsulfoxide-d6
ES-MS electrospray mass spectroscopy h hour, hours
1H NMR proton nuclear magnetic resonance
HPLC high performance liquid chromatography
J coupling constant (NMR spectroscopy)
LC liquid chromatography
LC-MS liquid chromatography-mass spectrometry m multiplet
M molar
MeCN acetonitrile
MeOH methanol
Me4Si tetramethylsilane min minute, minutes
MS mass spectrum, mass spectrometry m/z mass-to-charge ratio
N normal
NMP N-methylpyrrolidine
PS-DIEA polystyrene-bound diisopropylethylamine
Rf retention factor (TLC)
RT retention time (HPLC) rt room temperature s singlet t triplet
THF tetrahydrofuran
TFA trifluoroacetic acid
TLC thin layer chromatography v/v volume per volume w/v weight per volume w/w weight per weight.
[050] The compounds described in the Examples are intended to be representative of the invention, and it will be understood that the scope of the invention is not limited by the scope of the examples. Those skilled in the art will recognize that the invention may be practiced with variations on the disclosed structures, materials, compositions and methods, and such variations are regarded as within the ambit of the invention.
[051] Example 1
Preparation of tert-butyl 2-cvclopropyl-4-(3,3,3-trifluoropropoxyl-5,6,8,9-tetrahydro- 7H-pyrimido[4,5-d]azepine-7-carboxylate
Figure imgf000016_0001
[052] Step 1 : Preparation of tert-butyl 2-cvclopropyl-4-hvdroxy-5,6,8,9-tetrahydro-7H - pyrimido[4,5-d]azepine-7-carboxylate
Figure imgf000016_0002
[053] Commercially available 1- tert-butyl 4-ethyl 5-oxoazepane-1 ,4-dicarboxylate (6.0 g, 21.0 mmol) and cyclopropanecarboximidamide hydrochloride (2.54 g, 21.0 mmol) were combined and dissolved in ethyl alcohol (21 mL ). Sodium ethoxide (21 weight % solution in ethyl alcohol) (7.85 mL , 21.0 mmol) was added, and the mixture was heated at 80°C for 2 h. The contents of the reaction flask were cooled to 0°C and filtered. The solids were washed with cold methanol and dried in a 40°C vacuum oven to provide the title compound as an off-white solid (4.6 g, 72%). 1H NMR (300 MHz, DMSO-d6) δ 12.55 (s, 1 H), 3.46-3.36 (m, 4 H), 2.71-2.63 (m, 4 H), 1.88-1.79 (m, 1 H), 1.38 (s, 9 H), 0.95-0.93 (m, 4 H); ES-MS m/z 306.2 [M+ H+]; HPLC RT (min) 2.34. [054] Step 2: Preparation of tert-butyl 4-chloro-2-cvclopropyl-5,6,8,9-tetrahydro-7H - pyrimidof4,5-cnazepine-7-carboxylate
Figure imgf000017_0001
[055] tert-Butyl 2-cyclopropyl-4-hydroxy-5,6,8,9-tetrahydro-7H-pyrimido[4,5-d]azepine-7- carboxylate (4.61 g, 15.1 mmol) was dissolved in phosphorus oxychloride (14 mL) and heated to 90°C. Λ/,Λ/-Dimethylaniline (1.91 mL, 15.1 mmol) was added, and heating was continued for 1 h. The solution was cooled to rt, poured onto ice, and stirred for 3 h. The aqueous mixture was treated with 5N aqueous sodium hydroxide solution until the mixture reached a stable pH 10. The basic solution was extracted with chloroform / isopropyl alcohol (4:1) (3x). The organic extracts were combined, dried (MgSO4), and concentrated under reduced pressure. The resulting crude orange oil was purified by flash chromatography on silica gel eluted first with 1 :1 hexanes / ethyl acetate followed by 10% 2M solution of ammonia in methanol / 90% 1 :1 hexanes / ethyl acetate. Fractions containing the chloride were combined and concentrated under reduced pressure. The chloride was dissolved in anhydrous tetrahydrofuran and stirred with di-tert-butyl- dicarbonate (3.29 g, 15.1 mmol) for 1 h. The reaction mixture was concentrated under reduced pressure to remove solvent and was purified by flash chromatography on silica gel eluted with 9:1 hexanes / ethyl acetate. The title compound was collected as a pale yellow oil that crystallized upon standing (3.3 g, 67%). 1H NMR (300 MHz, CDCI3) δ 3.62- 3.57 (m, 4 H), 3.09-3.04 (m, 4 H), 2.18-2.10 (m, 1 H), 1.48 (s, 9 H), 1.13-1.00 (m, 4 H); ES-MS m/z 324.1 [M+H+]; HPLC RT (min) 3.46.
[056] Step 3: Preparation of tert-butyl 2-cvclopropyl-4-(3,3,3-trifluoropropoxy)-5,6,8,9- tetrahydro-7H-pyrimido[4,5-d]azepine-7-carboxylate
Figure imgf000017_0002
[057] 3,3,3-Trifluoropropan-1-ol (0.43 g, 3.7 mmol) was dissolved in 1 ,4-dioxane (5 mL). Potassium tert-butoxide (572 mg, 5.1 mmol) was added, and the mixture was stirred at rt for 15 min. tert-Butyl 4-chloro-2-cyclopropyl-5,6,8,9-tetrahydro-7H-pyrimido[4,5- d]azepine-7-carboxylate (1.1 g, 3.4 mmol) was then added as a solution in 1 ,4-dioxane (5 ml_). The mixture was heated at 50°C overnight. The resulting white suspension was concentrated under reduced pressure, and the residue was partitioned between water and ethyl acetate. The organic layer was separated, dried (MgSO4), and concentrated under reduced pressure to give the title compound as a colorless oil (1.2 g, 94%). 1H NMR (300 MHz, CD3OD) δ 4.56 (t, 2 H), 3.64-3.48 (m, 4 H), 3.05-3.01 (m, 2 H), 2.90-2.86 (m, 2 H), 2.76-2.61 (m, 2 H), 2.11-2.01 (m, 1 H), 1.45 (s, 9 H), 1.09-0.97 (m, 4 H); LC-MS m/z 402.0 [M+H+]; HPLC RT (min) 2.69.
[058] Example 2
Preparation of 2-cvclopropyl-4-(3,3,3-trifluoropropoxy)-6,7,8,9-tetrahvdro-5H- pyrimido[4,5-d]azepine dihvdrochloride
Figure imgf000018_0001
[059] tert-Butyl 2-cyclopropyl-4-(3,3,3-trifluoropropoxy)-5,6,8,9-tetrahydro-7H- pyrimido[4,5-d]azepine-7-carboxylate (Example 1 , 1.2 g, 3.0 mmol) was dissolved in 1 ,4- dioxane (3 mL) and treated with a solution of 4N HCI in 1 ,4-dioxane (3.4 mL, 13.6 mmol). The mixture was stirred at rt overnight resulting in a white suspension. The product was collected by filtration to provide the title compound as a white solid (0.80 g, 59%). 1H NMR (300 MHz,CD3OD) δ 4.77 (t, 2 H), 3.58-3.53 (m, 2 H), 3.49-3.41 (m, 4 H), 3.25-3.21 (m, 2 H), 2.88-2.73 (m, 2 H), 2.37-2.30 (m, 1 H), 1.47-1.42 (m, 4 H); LC-MS m/z 302.3 [M+H+]; HPLC RT (min) 1.64.
[060] Example 3
Preparation of 2-cyclopropyl-4-propyl-6,7,8,9-tetrahydro-5H-pyrimido[4,5-d]azepine hydrochloride
Figure imgf000018_0002
[061] tert-Butyl 4-Ghloro-2-cyclopropyl-5,6,8,9-tetrahyclro-7H-pyrimiclo[4,5-d]azepine-7- carboxylate (70 mg, 0.22 mmol) was dissolved in a solution of 10:1 THF:NMP. The solution was degassed and purged with nitrogen twice before adding iron(III) acetylacetonate (4 mg, 0.011 mmol). The reaction solution was then slowly treated with N-propylmagnesium bromide (160 μl_, 0.32 mmol). After 20 min, the reaction was diluted with diethyl ether and water. The organic layer was separated, dried over MgSO4, and concentrated to an amber oil that analyzed as tert-butyl 2-cyclopropyl-4-propyl-5, 6,8,9- tetrahydro-7H-pyrimido[4,5-d]azepine-7-carboxylate. 1H NMR (300 MHz, CD3OD) δ 3.64- 3.54 (m, 4 H), 3.07-3.03 (m, 2 H), 2.96-2.92 (m, 2 H), 2.76-2.71 (m, 2 H), 2.12-2.03 (m, 1 H), 1.72-1.59 (m, 2 H), 1.42 (bs, 9 H), 1.18-0.95 (m, 7 H); LC-MS m/z 332.2 [M+H+]; HPLC RT (min) 2.92.
[062] tert-Butyl 2-cyclopropyl-4-propyl-5,6,8,9-tetrahydro-7H-pyrimido[4,5-d]azepine-7- carboxylate was dissolved in 1 ,4-dioxane (1 mL) and treated with a solution of 4N HCI in 1 ,4-dioxane (1 mL) and stirred at rt until TLC indicated all starting material had been consumed. The reaction mixture was concentrated under reduced pressure. The resulting oil was taken up in acetonitrile and concentrated by rotary evaporation until a solid was obtained. The solids were collected by filtration and rinsed with acetonitrile to give 41 mg of the title compound as a tan solid (57%). 1H NMR (300 MHz, CD3OD) δ 3.48-3.41 (m, 4 H), 3.36-3.32 (m, 4 H), 3.02-2.97 (m, 2 H), 2.42-2.33 (m, 1 H), 1.87-1.68 (m, 2 H), 1.44-1.38 (m, 4 H), 1.08 (t, 3 H); LC-MS m/z232.3 [M+H+]; HPLC RT (min) 0.35.
[063] Example 4
Preparation of 2-cvclopropyl-9-methyl-4-(3,3,3-trifluoropropoxy)-6,7,8,9- tetrahvdro-5H-pyrimido[4,5-d]azepine dihydrochloride
Figure imgf000019_0001
[064] Step 1 : Preparation of 1-tert-butyl 4-ethyl 6-methyl-5-oxoazepane-1 ,4- dicarboxylate
Figure imgf000020_0001
[065] Sodium hydride, 95% (186 mg, 7.36 mmol) was suspended in THF (10 ml_). The suspension was cooled to 0°C, and 1- tert-butyl 4-ethyl-5-oxoazepane-1 ,4-dicarboxylate (1.0 g, 3.5 mmol) in THF (5 mL ) was added. The mixture was allowed to warm to rt and was stirred for an additional 30 min. The contents of the flask were cooled to -78°C, and n-BuLi (1.4 ml_, 3.5 mmol) was added dropwise. Once the addition was complete, the solution was stirred for another 10 min at -78°C. lodomethane (240 μL, 3.85 mmol) was added, and the mixture was allowed to return to rt. An excess of water was added, and the mixture was extracted with ethyl acetate. The organic layer was separated, dried over anhydrous sodium sulfate, and concentrated in vacuo. The resulting crude material was purified using Biotage® flash 4OS (2:1 hexanes / ethyl acetate) to afford 250 mg (24%) of the desired product. 1H NMR (300 MHz, CDCI3) δ 4.07-4.14 (m, 3 H), 3.58-3.69 (m, 2 H), 2.72-2.90 (m, 3 H), 1.95 (br s, 2 H), 1.36 (s, 9 H), 1.16-1.24 (m, 3 H), 1.06 (d, 3 H); ES-MS m/z 300.2 [M+H+]; HPLC RT (min) 2.97.
[066] Step 2: Preparation of tert-butyl 2-cvclopropyl-4-hvdroxy-9-methyl-5,6,8,9- tetrahvdro-7/-/-pyrimido[4,5-d]azepine-7-carboxylate
Figure imgf000020_0002
[067] Sodium ethoxide solution (21 weight % ethanol, 2.24 mL, 1.45 mmol) was added dropwise to a solution of 1- tert-butyl 4-ethyl 6-methyl-5-oxoazepane-1,4-dicarboxylate (290 mg, 0.97 mmol) in ethanol (6 mL). Cyclopropylcarbamidine hydrochloride (175 mg, 1.45 mmol) was added, and the mixture was heated at reflux for 3 h. The contents of the flask were cooled to rt, and the solvent was removed under reduced pressure. The residue was suspended in water and extracted with chloroform (2x). The organic layers were combined and concentrated to afford 200 mg (64%) of crude product, which was used in the next step without further purification. 1H NMR (300 MHz, CDCI3) δ 4.01-4.06 (m, 1 H), 3.37-3.67 (m, 4 H), 2.74-3.00 (m, 4 H), 1.84 (br s, 1 H), 1.44 (s, 9 H), 1.18 (d, 1 H), 0.78-0.92 (m, 4 H); ES-MS m/z 320.2 [M+H+]; HPLC RT (min) 2.50.
[068] Step 3: Preparation of 4-chloro-2-cvclopropyl-9-methyl-6,7.8,9-tetrahydro-5H- pyrimido[4,5-d]azepine
Figure imgf000021_0001
[069] tert-Butyl 2-cyclopropyl-4-hydroxy-9-methyl-5,6,8,9-tetrahydro-7H-pyrimido[4,5- d]azepine-7-carboxylate (200 mg, 0.63 mmol) was suspended in phosphorus oxychloride (0.6 ml_, 6.3 mmol) and heated to 80°C. Once at 80°C, N,N-dimethylaniline (0.04 mL, 0.31 mmol) was added, and the reaction mixture was heated for an additional hour. The mixture was then cooled to rt, poured into ice, and stirred for 1 h. The aqueous solution was brought to pH 8-9 by addition of 2N aqueous sodium hydroxide solution. The basic aqueous solution was extracted with chloroform (2x), dried over anhydrous sodium sulfate, and concentrated in vacuo to afford 80 mg of desired product (54%). 1H NMR (300 MHz, CD3OD) δ 3.28-3.36 (m, 2 H), 3.11-3.30 (m, 2 H), 2.76-2.82 (m, 2 H), 2.15- 2.20 (m, 1 H), 1.45 (br s, 1 H), 1.34 (d, 3 H), 0.94-1.10 (m, 4 H); ES-MS m/z 238.4 [M+H+]; HPLC RT (min) 1.40.
[070] Step 4: Preparation of tert-butyl 4-chloro-2-cvclopropyl-9-methyl-5,6,8,9- tetrahvdro-7H-pyrimido[4,5-d]azepine-7-carboxylate
Figure imgf000021_0002
[071] To a solution of 4-chloro-2-cyclopropyl-9-methyl-6,7,8,9-tetrahydro-5H- pyrimido[4,5-d]azepine (60 mg, 0.25 mmol) in THF (2 mL) was added water (0.5 mL), sodium bicarbonate (23 mg, 0.28 mmol), and di- tert-butyl dicarbonate (61 mg, 0.28 mmol). The mixture was stirred at rt for 16 h, then was diluted with water, and extracted with ethyl acetate. The organic layer was separated, dried over anhydrous sodium sulfate, and concentrated in vacuo. The resulting crude material was purified using Biotage® flash 4OS (3:1 hexanes / ethyl acetate) to afford 38 mg (45%) of the desired product. 1H NMR (300 MHz, CD3OD) δ 3.50-3.70 (m, 2 H), 3.41-3.49 (m, 1 H), 3.32-3.40 (m, 2 H), 3.14-3.42 (m, 2 H), 2.18-2.32 (m, 1 H), 1.52 (s, 9 H), 1.32 (d, 3 H), 0.93-1.08 (m, 4 H); ES-MS m/z 338.1 [M+H+]; HPLC RT (min) 3.75.
[072] Step 5: Preparation of 2-cvclopropyl-9-methyl-4-(3.3,3-trifluoropropoxy)-6,7,8,9- tetrahvdro-5H-pyrimido[4,5-d]azepine dihvdrochloride
Figure imgf000022_0001
[073] To a solution of 3,3,3-trifluoropropan-1-ol (14 mg, 0.12 mmol) in 1 ,4-dioxane (1 ml.) was added potassium tert-butoxide (58 mg, 0.52 mmol), and the resulting mixture was stirred at rt for 15 min. tert-Butyl 4-chloro-2-cyclopropyl-9-methyl-5,6,8,9-tetrahydro- 7H-pyrimido[4,5-d]azepine-7-carboxylate (35 mg, 0.10 mmol) was then added, and the mixture was heated at 80°C overnight. The reaction was cooled to rt and filtered through a pad of Celite®. The filtrate was concentrated under reduced pressure, and the crude material was re-dissolved in 1 ,4-dioxane. A 4M solution of hydrochloric acid in dioxane (0.1 ml_, 0.4 mmol) was added, and the mixture was stirred for 3 h. The dioxane solvent was removed under reduced pressure, and the crude residue was triturated with ethyl acetate to afford 12 mg (29%) of desired product. 1H NMR (300 MHz, CD3OD) δ 4.75 (t, 2 H), 3.61-3.71 (m, 1 H), 3.41-3.62 (m, 4 H), 3.12-3.34 (m, 2 H), 2.77-2.83 (m, 2 H), 2.36- 2.42 (m, 1 H), 1.57 (d, 3 H), 1.38-1.50 (m, 5 H); ES-MS m/z 316.2 [M+H+]; HPLC RT (min) 1.90.
[074] Example 5
Preparation of 2-cvclopropyl-7-methyl-4-(3,3,3-trifluoropropoxy)-6,7,8,9-tetrahvdro-
5H-pyrimido[4,5-d]azepine
Figure imgf000022_0002
[075] 2-Cyclopropyl-4-(3,3,3-trifluoropropoxy)-6,7,8,9-tetrahydro-5H-pyrimido[4,5- d]azepine dihydrochloride of Example 2 (92 mg, 0.3 mmol) was suspended in ethyl acetate and washed with saturated aqueous sodium bicarbonate solution. The organic layer was separated, dried (MgSO4), and concentrated under reduced pressure to a colorless oil. The oil was dissolved in anhydrous DMF (0.5 mL) and added to a suspension of 95% sodium hydride (8 mg, 0.3 mmol) in DMF (0.5 mL). The mixture was stirred for 15 min at ambient temperature, lodomethane (20 μL , 0.3 mmol) was added, and the solution was stirred for an additional 4 h. The contents of the flask were partitioned between ethyl acetate and water. The organic layer was separated and concentrated to a crude oil under reduced pressure. The resulting crude was purified by HPLC eluted with an acetonitrile / water mixture to provide the title compound as a yellow solid (20 mg, 27%). 1H NMR (300 MHz, CD3OD) δ 4.55 (t, 2 H), 3.02-2.99 (m, 2 H), 2.90- 2.86 (m, 2 H), 2.73-2.65 (m, 2 H), 2.64-2.60 (m, 2 H), 2.55-2.51 (m, 2 H), 2.37 (s, 3 H), 2.10-2.01 (m, 1 H), 1.06-0.97 (m, 4 H); ES-MS m/z316.2 [M+H+]; HPLC RT (min) 1.71.
[076] Example 6
Preparation of 7-acetyl-2-cvclopropyl-4-(3,3,3-trifluoropropoxy) -6,7,8,9-tetrahydro-
5H-pyrimido[4,5-d]azepine
Figure imgf000023_0001
[077] 2-Cyclopropyl-4-(3,3,3-trifluoropropoxy)-6,7,8,9-tetrahydro-5H-pyrimido[4,5- d]azepine dihydrochloride of Example 2 (92 mg, 0.3 mmol) was suspended in ethyl acetate and washed with saturated aqueous sodium bicarbonate solution. The organic layer was separated, dried (MgSO4), and concentrated under reduced pressure to a colorless oil. The oil was dissolved in anhydrous dichloromethane (1 mL) and triethylamine (0.13 mL, 0.9 mmol) and treated with acetyl chloride (30 μL, 0.45 mmol). The solution was stirred at ambient temperature for 4 h. The contents of the flask were partitioned between ethyl acetate and water. The organic layer was separated and concentrated to a crude oil under reduced pressure. The resulting crude was purified by HPLC eluted with an acetonitrile / water mixture to provide the title compound as a yellow solid (82 mg, 99%). 1H NMR (300 MHz, CD3OD) δ 4.59-4.53 (m, 2 H), 3.76-3.64 (m, 4 H), 3.13-3.09 (m, 1 H), 3.04-3.00 (m, 1 H), 2.98-2.94 (m, 1 H), 2.89-2.85 (m, 1 H), 2.75- 2.60 (m, 2 H), 2.15 (d, 3 H), 2.10-2.02 (m, 1 H), 1.07-0.97 (m, 4 H); ES-MS m/z 344.3 [M+H+]; HPLC RT (min) 2.07. [078] Using the methods described for Examples 1 through 6, and using the appropriate starting materials, the compounds of Table 1 were prepared in analogous manner.
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Table 1
Figure imgf000030_0001
Figure imgf000031_0001
Table 1
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Table 1
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Table 1
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Table 1
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Table 1
Figure imgf000090_0001
Figure imgf000091_0001
Table 1
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Methods of Use
[079] As used herein, various terms are defined below.
[080] When introducing elements of the present invention or the preferred embodiment(s) thereof, the articles "a," "an," "the," and "said" are intended to mean that there are one or more of the elements. The terms "comprising," "including," and "having" are intended to be inclusive and mean that there may be additional elements other than the listed elements.
[081] The term "subject" as used herein includes mammals (e.g., humans and animals).
[082] The term "treatment" includes any process, action, application, therapy, or the like, wherein a subject, including a human being, is provided medical aid with the object of improving the subject's condition, directly or indirectly, or slowing the progression of a condition or disorder in the subject.
[083] The term "combination therapy" or "co-therapy" means the administration of two or more therapeutic agents to treat a disease, condition, and/or disorder. Such administration encompasses co-administration of two or more therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each inhibitor agent. In additioi such administration encompasses use of each type of therapeutic agent in a sequential manner.
[084] The phrase "therapeutically effective" means the amount of each agent administered that will achieve the goal of improvement in a disease, condition, and/or disorder severity, while avoiding or minimizing adverse side effects associated with the given therapeutic treatment.
[085] The term "pharmaceutically acceptable" means that the subject item is appropriate for use in a pharmaceutical product.
[086] The compounds of Formula (I) of this invention are expected to be valuable as therapeutic agents. Accordingly, an embodiment of this invention includes a method of treating the various conditions in a patient (including mammals) which comprises administering to said patient a composition containing an amount of the compound of Formula (I) that is effective in treating the target condition.
[087] The compounds of the present invention interact with the 5-HT2C receptor and may be used in the treatment or prevention of diseases and/or behaviors that involve the 5-HT2C receptor. These diseases and/or behaviors include, for example, obesity, obesity related disorders such as diabetes, feeding behavior, eating disorders such as bulimia, anorexia nervosa and premenstrual tension.
[088] For example, an object of this invention is to provide methods for treating obesity and inducing weight loss in an individual by administration of a compound of the invention. The method of the invention comprises administering to an individual a therapeutically effective amount of at least one compound of the invention, or a prodrug thereof, which is sufficient to induce weight loss. The invention further comprises a method of preventing weight gain in an individual by administering an amount of at least one compound of the invention, or a prodrug thereof, which is sufficient to prevent weight gain.
[089] Further diseases and/or behaviors which can be treated or prevented include central nervous disorders, depressions, anxiety disorders, obsessive-compulsive disorders, sleep disorders, sexual dysfunction, psychoses, migraine, schizophrenia, drug or alcohol addiction and chronic fatigue syndrome.
[090] The present invention also relates to the use of the compounds of this invention for the treatment of obesity-related diseases including associated dyslipidemia and other obesity- and overweight-related complications such as, for example, cholesterol gallstones, gallbladder disease, gout, cancer (e.g., colon, rectum, prostate, breast, ovary, endometrium, cervix, gallbladder, and bile duct), menstrual abnormalities, infertility, polycystic ovaries, osteoarthritis, and sleep apnea, as well as for a number of other pharmaceutical uses associated therewith, such as the regulation of appetite and food intake, dyslipidemia, hypertriglyceridemia, Syndrome X, type 2 diabetes (non-insulin- dependent diabetes), atherosclerotic diseases such as heart failure, hyperlipidemia, hypercholesteremia, low HDL levels, hypertension, cardiovascular disease (including atherosclerosis, coronary heart disease, coronary artery disease, and hypertension), cerebrovascular disease such as stroke, and peripheral vessel disease. The compounds of this invention may also be useful for treating physiological disorders related to, for example, regulation of insulin sensitivity, inflammatory response, plasma triglycerides, HDL, LDL and cholesterol levels and the like.
[091] Compounds of Formula (I) may be administered alone or in combination with one or more additional therapeutic agents. Combination therapy includes administration of a single pharmaceutical dosage formulation which contains a compound of Formula (I) and one or more additional therapeutic agents, as well as administration of the compound of Formula (I) and each additional therapeutic agents in its own separate pharmaceutical dosage formulation. For example, a compound of Formula (I) and a therapeutic agent may be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent may be administered in separate oral dosage formulations.
[092] Where separate dosage formulations are used, the compound of Formula (I) and one or more additional therapeutic agents may be administered at essentially the same time (e.g., concurrently) or at separately staggered times (e.g., sequentially).
[093] For example, the compound of Formula (I) may be used in combination with other therapies and drugs useful for the treatment of obesity. For example, anti-obesity drugs include β-3 adrenergic receptor agonists such as CL 316,243; cannabinoid (e.g., CB-1) antagonists such as Rimonabant; neuropeptide-Y receptor antagonists; neuropeptide Y5 inhibitors; apo-B/MTP inhibitors; 11 β-hydroxy steroid dehydrogenase-1 inhibitors; peptide YY3-36 or analogs thereof; MCR4 agonists; CCK-A agonists; monoamine reuptake inhibitors; sympathomimetic agents; dopamine agonists; melanocyte-stimulating hormone receptor analogs; melanin concentrating hormone antagonists; leptin; leptin analogs; leptin receptor agonists; galanin antagonists; lipase inhibitors; bombesin agonists; thyromimetic agents; dehydroepiandrosterone or analogs thereof; glucocorticoid receptor antagonists; orexin receptor antagonists; ciliary neurotrophic factor; ghrelin receptor antagonists; histamine-3 receptor antagonists; neuromedin U receptor agonists; appetite suppressants, such as, for example, sibutramine (Meridia); and lipase inhibitors, such as, for example, orlistat (Xenical). The compounds of the present invention may also be administered in combination with a drug compound that modulates digestion and/or metabolism such as drugs that modulate thermogenesis, lipolysis, gut motility, fat absorption, and satiety.
[094] In addition, the compound of Formula (I) may be administered in combination with one or more of the following agents for the treatment of diabetes or diabetes-related disorders including PPAR ligands (agonists, antagonists), insulin secretagogues, for example, sulfonylurea drugs and non-sulfonylurea secretagogues, α-glucosidase inhibitors, insulin sensitizers, hepatic glucose output lowering compounds, and insulin and insulin derivatives. Such therapies may be administered prior to, concurrently with, or following administration of the compounds of the invention. Insulin and insulin derivatives include both long and short acting forms and formulations of insulin. PPAR ligands may include agonists and/or antagonists of any of the PPAR receptors or combinations thereof. For example, PPAR ligands may include ligands of PPAR-α, PPAR-γ, PPAR-δ or any combination of two or three of the receptors of PPAR. PPAR ligands include, for example, rosiglitazone, troglitazone, and pioglitazone. Sulfonylurea drugs include, for example, glyburide, glimepiride, chlorpropamide, tolbutamide, and glipizide, α- glucosidase inhibitors that may be useful in treating diabetes when administered with a compound of the invention include acarbose, miglitol, and voglibose. Insulin sensitizers that may be useful in treating diabetes include PPAR-γ agonists such as the glitazones (e.g., troglitazone, pioglitazone, englitazone, MCC-555, rosiglitazone, and the like) and other thiazolidinedione and non-thiazolidinedione compounds; biguanides such as metformin and phenformin; protein tyrosine phosphatase- 1 B (PTP-1B) inhibitors; dipeptidyl peptidase IV (DPP-IV) inhibitors, and 11 beta-HSD inhibitors. Hepatic glucose output lowering compounds that may be useful in treating diabetes when administered with a compound of the invention include glucagon anatgonists and metformin, such as Glucophage and Glucophage XR. Insulin secretagogues that may be useful in treating diabetes when administered with a compound of the invention include sulfonylurea and non-sulfonylurea drugs: GLP-1 , GIP, PACAP, secretin, and derivatives thereof; nateglinide, meglitinide, repaglinide, glibenclamide, glimepiride, chlorpropamide, glipizide. GLP-1 includes derivatives of GLP-1 with longer half-lives than native GLP-1 , such as, for example, fatty-acid derivatized GLP-1 and exendin.
[095] Compounds of the invention may also be used in methods of the invention in combination with drugs commonly used to treat lipid disorders in patients. Such drugs include, but are not limited to, HMG-CoA reductase inhibitors, nicotinic acid, fatty acid lowering compounds (e.g., acipimox); lipid lowering drugs (e.g., stanol esters, sterol glycosides such as tiqueside, and azetidinones such as ezetimibe), ACAT inhibitors (such as avasimibe), bile acid sequestrants, bile acid reuptake inhibitors, microsomal triglyceride transport inhibitors, and fibric acid derivatives. HMG-CoA reductase inhibitors include, for example, lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin, rivastatin, itavastatin, cerivastatin, and ZD-4522. Fibric acid derivatives include, for example, clofibrate, fenofibrate. bezafibrate, ciprofibrate, beclofibrate, etofibrate, and gemfibrozil. Sequestrants include, for example, cholestyramine, colestipol, and dialkylaminoalkyl derivatives of a cross-linked dextran.
[096] Compounds of the invention may also be used in combination with anti¬ hypertensive drugs, such as, for example, β-blockers and ACE inhibitors. Examples of additional anti-hypertensive agents for use in combination with the compounds of the present invention include calcium channel blockers (L-type and T-type; e.g., diltiazem, verapamil, nifedipine, amlodipine and mybefradil), diuretics (e.g., chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzthiazide, ethacrynic acid tricrynafen, chlorthalidone, furosemide, musolimine, bumetanide, triamtrenene, amiloride, spironolactone), renin inhibitors, ACE inhibitors (e.g., captopril, zofenopril, fosinopril, enalapril, ceranopril, cilazopril, delapril, pentopril, quinapril, ramipril, lisinopril), AT-1 receptor antagonists (e.g., losartan, irbesartan, valsartan), ET receptor antagonists (e.g., sitaxsentan, atrsentan, neutral endopeptidase (NEP) inhibitors, vasopepsidase inhibitors (dual NEP-ACE inhibitors) (e.g., omapatrilat and gemopatrilat), and nitrates.
[097] The compound of Formula (I) may also be utilized, in free base form or in compositions, as well as in research and diagnostics or as analytical reference standards, and the like, which are well known in the art. Therefore, the present invention includes compositions which are comprised of an inert carrier and an effective amount of a compound of Formula (I), or a salt, or ester thereof. An inert carrier is any material which does not interact with the compound to be carried and which lends support, means of conveyance, bulk, traceable material, and the like to the compound to be carried. An effective amount of the compound is that amount which produces a result or exerts an influence on the particular procedure being performed.
[098] It is anticipated that prodrug forms of the compounds of this invention will prove useful in certain circumstances, and such compounds are also intended to fall within the scope of the invention. Prodrug forms may have advantages over the parent compounds exemplified herein, in that they are better absorbed, better distributed, more readily penetrate the central nervous system, are more slowly metabolized or cleared, etc. Prodrug forms may also have formulation advantages in terms of crystallinity or water solubility. For example, compounds of the invention having one or more hydroxyl groups may be converted to esters or carbonates bearing one or more carboxyl, hydroxyl or amino groups, which are hydrolyzed at physiological pH values or are cleaved by endogenous esterases or lipases in vivo (see, e.g., U.S. Patent Nos. 4,942,184; 4,960,790; 5,817,840; and 5,824,701 , all of which are incorporated herein by reference in their entirety, and references therein).
Pharmaceutical Compositions
[099] Based on the above tests, or other well known assays used to determine the efficacy for treatment of conditions identified above in mammals, and by comparison of these results with the results of known medicaments that are used to treat these conditions, the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication. The amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
[100] The total amount of the active ingredient to be administered may generally range from about 0.001 mg/kg to about 200 mg/kg. A unit dosage may contain from about 0.05 mg to about 1500 mg of active ingredient, and may be administered one or more times per day. The daily dosage for administration by injection, including intravenous, intramuscular, subcutaneous, and parenteral injections, and use of infusion techniques may be from about 0.01 to about 200 mg/kg. The daily rectal dosage regimen may be from 0.01 to 200 mg/kg of total body weight. The transdermal concentration may be that required to maintain a daily dose of from 0.01 to 200 mg/kg.
[101] Of course, the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age of the patient, the diet of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like. The desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt thereof may be ascertained by those skilled in the art using conventional treatment tests.
[102] The compounds of this invention may be utilized to achieve the desired pharmacological effect by administration to a subject in need thereof in an appropriately formulated pharmaceutical composition. A subject, for example, may be a mammal, including a human, in need of treatment for a particular condition or disease. Therefore, the present invention includes pharmaceutical compositions which are comprised of a pharmaceutically acceptable carrier and a pharmaceutically effective amount of a compound identified by the methods described herein, or a pharmaceutically acceptable salt or ester thereof. A pharmaceutically acceptable carrier is any carrier which is relatively non-toxic and innocuous to a patient at concentrations consistent with effective activity of the active ingredient so that any side effects ascribable to the carrier do not vitiate the beneficial effects of the active ingredient. A pharmaceutically effective amount of a compound is that amount which produces a result or exerts an influence on the particular condition being treated. The compounds identified by the methods described herein may be administered with a pharmaceutically-acceptable carrier using any effective conventional dosage unit forms, including, for example, immediate and timed release preparations, orally, parenterally, topically, or the like. [103] For oral administration, the compounds may be formulated into solid or liquid preparations such as, for example, capsules, pills, tablets, troches, lozenges, melts, powders, solutions, suspensions, or emulsions, and may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions. The solid unit dosage forms may be a capsule which can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and corn starch.
[104] In another embodiment, the compounds of this invention may be tableted with conventional tablet bases such as lactose, sucrose, and cornstarch in combination with binders such as acacia, cornstarch, or gelatin; disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum; lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example, talc, stearic acid, or magnesium, calcium or zinc stearate; dyes; coloring agents; and flavoring agents intended to enhance the aesthetic qualities of the tablets and make them more acceptable to the patient. Suitable excipients for use in oral liquid dosage forms include diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both.
[105] Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example, those sweetening, flavoring and coloring agents described above, may also be present.
[106] The pharmaceutical compositions of this invention may also be in the form of oil- in-water emulsions. The oily phase may be a vegetable oil such as liquid paraffin or a mixture of vegetable oils. Suitable emulsifying agents may be naturally occurring gums, naturally occurring phosphatides, esters or partial esters derived from fatty acids and hexitol anhydrides, and condensation products of said partial esters. The emulsions may also contain sweetening and flavoring agents. [107] Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil such or in a mineral oil. The oily suspensions may contain a thickening agent. The suspensions may also contain one or more preservatives, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents.
[108] Syrups and elixirs may be formulated with sweetening agents such as, for example, glycerol, propylene glycol, sorbitol, or sucrose. Such formulations may also contain a demulcent, and preservative, flavoring and coloring agents.
[109] The compounds of this invention may also be administered parenterally, that is, subcutaneously, intravenously, intramuscularly, or interperitoneally, as injectable dosages of the compound in a physiologically acceptable diluent with a pharmaceutical carrier, with or without the addition of a pharmaceutically acceptable surfactants, suspending agents, emulsifying agents or other pharmaceutical adjuvants.
[110] The parenteral compositions of this invention may typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimize or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulation ranges from about 5% to about 15% by weight. The surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB.
[111] The pharmaceutical compositions may be in the form of sterile injectable aqueous suspensions. Such suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents.
[112] The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Diluents and solvents that may be employed are, for example, water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile fixed oils are conventionally employed as solvents or suspending media. For this purpose, any bland, fixed oil may be employed including synthetic mono or diglycerides. In addition, fatty acids such as oleic acid may be used in the preparation of injectables.
[113] A composition of the invention may also be administered in the form of suppositories for rectal administration of the drug. These compositions may be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such material are, for example, cocoa butter and polyethylene glycol.
[114] Another formulation employed in the methods of the present invention employs transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art (see, e.g., U.S. Patent No. 5,023,252, incorporated herein by reference). Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
[115] It may be desirable or necessary to introduce the pharmaceutical composition to the patient via a mechanical delivery device. The construction and use of mechanical delivery devices for the delivery of pharmaceutical agents is well known in the art. For example, direct techniques for administering a drug directly to the brain usually involve placement of a drug delivery catheter into the patient's ventricular system to bypass the blood-brain barrier. One such implantable delivery system, used for the transport of agents to specific anatomical regions of the body, is described in U.S. Patent No. 5,011 ,472, incorporated herein by reference.
[116] The compositions of the invention may also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired. Any of the compositions of this invention may be preserved by the addition of an antioxidant such as ascorbic acid or by other suitable preservatives. Conventional procedures for preparing such compositions in appropriate dosage forms can be utilized.
[117] The compounds identified by the methods described herein may be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutical agents where the combination causes no unacceptable adverse effects. For example, the compounds of this invention can be combined with known anti-obesity, or with known antidiabetic or other indication agents, and the like, as well as with admixtures and combinations thereof.
[118] The compounds identified by the methods described herein may also be utilized, in free base form or in compositions, in research and diagnostics, or as analytical reference standards, and the like. Therefore, the present invention includes compositions which are comprised of an inert carrier and an effective amount of a compound identified by the methods described herein, or a salt or ester thereof. An inert carrier is any material which does not interact with the compound to be carried and which lends support, means of conveyance, bulk, traceable material, and the like to the compound to be carried. An effective amount of compound is that amount which produces a result or exerts an influence on the particular procedure being performed.
[119] Formulations suitable for subcutaneous, intravenous, intramuscular, and the like; suitable pharmaceutical carriers; and techniques for formulation and administration may be prepared by any of the methods well known in the art (see, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 20th edition, 2000)
[120] The structures, materials, compositions, and methods described herein are intended to be representative examples of the invention, and it will be understood that the scope of the invention is not limited by the scope of the examples. Those skilled in the art will recognize that the invention may be practiced with variations on the disclosed structures, materials, compositions and methods, and such variations are regarded as within the ambit of the invention.
Evaluation of Biological Activity
[121] In order that this invention may be better understood, the following examples are set forth. These examples are for the purpose of illustration only, and are not to be construed as limiting the scope of the invention in any manner. All publications mentioned herein are incorporated by reference in their entirety.
[122] Demonstration of the activity of the compounds of the present invention may be accomplished through in vitro, ex vivo, and in vivo assays that are well known in the art. For example, to demonstrate the activity of the compounds of the present invention, the following assays may be used.
In Vitro Assays
[123] Activity of the compounds of Formula (I) may be determined by measurement of their binding affinity and agonist activity at the 5-HT2C receptor. The following assays were performed to determine the effect of the compounds of Formula (I) on the 5-HT2C receptor.
[124] Membranes prepared from an AV-12 cell line expressing human recombinant 5- HT2C receptor were used for the [125I]-DOI displacement assay (SPA format) to examine binding affinity of compounds of Formula (I). The 5-HT2C SPA binding assay was performed in a buffer containing 50 mM Tris (pH7.4), 10 mM MgCI2, 10 μM pargyline, 0.1% sodium ascorbate, 0.5 mM EDTA, and 0.1% BSA. All assay components were diluted in this buffer. [125I] DOI (NEN - Cat. No. NEX255) and wheat agglutinin SPA beads PEI treated- type B (RPNQ0004, Amersham, Piscataway, NJ) were used for this binding assay. In a 96-well flex plate, 200 pM of the radiolabel was added followed by the appropriately dosed compounds and references. Membranes (5 μg/well, 50 μl_) and 50 μl of the SPA beads (250 μg/well) were added to each well. The reaction plates were mixed on a plate shaker for 20-30 minutes. The beads were then allowed to settle for about 1 hour before they were counted in a Wallac Microbeta.
[125] The functional activity of compounds of Formula (I) was measured using a FLIPR calcium mobilization assay in cells transfected with recombinant receptor. AV-12 cells expressing 5-HT2C receptors were placed into 96-well black plates (5 x104 cells per well/ 100 μL DMEM medium. The DMEM medium contained 10% dialysis FBS, 2 mM L- glutamine, 20 mM Hepes, 1 mg/ml G418, and 1 % antibiotic. The cell plates are incubated overnight at 37°C, 5% of CO2. Following incubation, the cell culture medium was removed, and 160 μL of 1X loading buffer (1X Hank's buffer contains 20 mM Hepes and 2.5 mM probenecid dissolved in 0.5 N NaOH) was added with calcium dye (Calcium 3 Assay Kit, Molecular Devices, Inc) into the cell plates. The cells were incubated at 37°C, 5% CO2 for 1 hour. Thereafter, the cell plates and compound dilution plates were placed into the FLIPR instrument, and 40 μL of varying concentrations of test compound or reference compound (alpha-Me-5-HT) were added into the cell plates. Increases in intracellular calcium were recorded at 488 nm during 3 minutes with the FLIPR. Peak fluorescence counts minus basal counts from 1 sec to 180 sec time points were used to determine agonist activity. Iterative curve-fitting and parameter estimations were analyzed using a four-parameter logistic equation and a program that generates dose- response curves. The potency of the compound was expressed in terms of the concentration required to achieve half-maximal stimulation (EC50) while the efficacy of the ligand was expressed as the percentage of its maximal stimulation relative to the maximal stimulation observed with the alpha-Me-5-HT curve from the same plate (E/Emax).
In Vivo Assays
Evaluation of Compound's Efficacy on the Reduction of Food Intake in Lean Overnight Fasted Rats
Fasted-Refed Acute Feeding Assay
[126] The purpose of this protocol is to determine the effect of a single dose of an unknown compound on food consumption of lean overnight fasted rats. The fasted-refed rat model is frequently used in the field of obesity to identify compounds with potential for anorectic effects. This animal model has been successfully used in the identification and characterization of the efficacy profile of compounds that are or have been used in the management of body weight in obese humans (see, e.g., Balvet, et al., Gen. Pharmacol. 13:293-297, 1982; Θrignaschi, et al., Br. J. Pharmacol. 127:1190-1194, 1999; McTavish and Heel, Drug 43:713-733, 1992; Rowland, et al., Life Sci. 36:2295-2300, 1985).
[127] A typical study includes 60-80 male Wistar rats (n=10/treatment group) with an average body weight of approximately 280 g. Rats are kept in standard animal rooms under controlled temperature and humidity and a 12/12 light dark cycle. Rats are single- housed in suspended cages with a mesh floor. Water and food are continuously available unless the animals are being fasted for the study.
[128] The rats are fasted overnight during the dark phase (total of approx. 16-18 hr). The animal is dosed with an assigned treatment (2 mg/mL). One hour after dosing, pre- weighed food jars are returned to the cage. Food intake is recorded 30, 60, 90, 180, and 240 minutes and 24 hours post-food return. At each time point, spillage is returned to the food jar and then the food jars are weighed. The amount of food consumed is determined for each time point. Difference between treatment group is determined using appropriate statistical analysis.
Evaluation of Compound's Efficacy on the Reduction of Body Weight and Food and Water Consumption in Obese Zucker fa/fa Rats
Chronic Feeding Assay
[129] The purpose of this protocol is to determine the effect of chronic administration of an unknown compound on body weight and food and water consumption in obese Zucker (fa/fa) rats. Obese Zucker rats are frequently used in the determination of compound efficacy in the reduction of body weight. This animal model has been successfully used in the identification and characterization of the efficacy profile of compounds that are or have been used in the management of body weight in obese humans (see, e.g., Al- Barazanji, et al., Obes Res. 8:317-323, 2000; Assimacopoulos-Jeannet, et al., Am. J. Physiol. 260(2 Pt 2):R278-283, 1991 ; Dryden, et al., Horm. Metab. Res. 31 :363-366, 1999; Edwards and Stevens, Pharmacol. Biochem. Behav. 47:865-872, 1994; Grinker, et al., Pharmacol. Biochem. Behav. 12:265-275, 1980).
[130] A typical study includes 60-80 male Zucker (fa/fa) rats (approximatelylO/treatment group) with an average body weight of approximately 550 g. Rats are kept in standard animal rooms under controlled temperature and humidity and a 12/12 light/dark cycle. Water and food are continuously available. Rats are single-housed in large rat shoeboxes containing grid floor. Animals are adapted to the grid floors and sham-dosed with study vehicle for at least four days before the recording of two-days baseline measurement of body weight and 24 h food and water consumption. Rats are assigned to one of 6-8 treatment groups based upon their baseline body weight. The groups are set up so that the mean and standard error of the mean of body weight are similar.
[131] Animals are orally gavaged (2 ml_/kg) daily before the dark phase of the light/dark cycle for a pre-determined number of days (typically 6-14 days) with their assigned dose/compound. At this time, body weight, food and water consumption are measured. On the final day, body weight is measured and animals are euthanized by CO2 inhalation.
[132] Other embodiments of the invention will be apparent to the skilled in the art from a consideration of this specification or practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with the true scope and spirit of the invention being indicated by the following claims.

Claims

Claims
1) A compound of Formula (I)
Figure imgf000117_0001
(I) wherein R1 is
. (C1-C4)alkyl,
• phenyl optionally substituted with halo, or
• (C3-C6)cycloalkyl optionally substituted with (C1-C3)alkyl;
R2 is
• C1-C6 alkyl,
• halo
• piperidin-1-yl
• pyrrolidin-1-yl
• phenylpyrrolidin-1-yl optionally substituted on the phenyl ring with up to two groups selected from halo, (C1-C3)alkyl, (CrC3)alkoxy, or CF3,
• phenylpiperidin-1 -yl optionally substituted on the phenyl ring with up to two groups selected fromhalo or CF3,
• phenoxypiperidin-1 -yl optionally substituted on the phenyl ring with up to two groups selected from halo or CF3,
• phenylpiperazin-1 -yl optionally substituted on the phenyl ring with up to two groups selected from (C1-C3)alkyl, halo, CF3, or (C1-C3)alkoxy,
• -NR5R6 wherein R5 is H or (C1-C3)alkyl, and
R6 is (C1-C3)alkyl optionally substituted with
• indoly-2-yl optionally substituted with halo,
• phenyl optionally substituted with up to two groups selected from (C1-C3)alkoxy, or
• benzodioxan-6-yl, or
• OR7 wherein R7 is
• H,
• (C3-C6)cycloalkyl optionally substituted with (C1-C3)alkyl,
• 2,3-dihydro-inden-2-yl,
• tetrahydrofuran-yl, or
• (C1-C6)alkyl optionally substituted with
• F up to the perfluoro level,
• (C3-C6)cycloalkyl,
• (CrC3)alkoxy,
• — N[(C1-C3)alkyl]2,
• — NH[(C1-C3)alkyl],
• morpholin-1-yl,
• phenyl optionally substituted with up to two groups selected from CF3, halo, (C1-C3)alkyl, NO2, (C1-C3)alkoxy, or N[(C1-C3)alkyl]2,
• 2,3-dihydro-benzo[1 ,4]dioxin-2-yl, • 2,3-dihydro-1 H-indol-2-yl,
• tetrahydrofuran-yl,
• 2-oxo-2, 3-dihydro-benzoimidazol-1 -yl,
• 2-oxo-pyrrolidin-1-yl, or
• — N[(C1-C3)alkyl]phenyl, wherein the phenyl is optionally substituted with up to two groups selected from CF3, halo, (C1-C3)alkyl, (C1- C3)alkoxy, and — N[(C1-C3)alkyl]2;
R3 is H or (C1-C3)alkyl; and
R4 is H, methyl, -C(O)O-R8, or -C(O)R9; wherein
R8 is (C1-C4)alkyl or phenyl optionally substituted with halo, (C1-C3)aIkyI, (C1-C3)alkoxy, or CF3, and
R9 is (C1-C4)alkyl or phenyl optionally substituted with halo, (C1-C3)alkyl, (C1-C3)alkoxy, or CF3;
or pharmaceutically acceptable salts thereof.
2. The compound of claim 1 , wherein
R1 is
• (C1-C4)alkyl, or
• (C3-C6)cycloalkyl optionally substituted with (C1-C3)aIkyI;
R2 is
• C1-C6 alkyl,
• halo • phenylpyrrolidin-1-yl optionally substituted on the phenyl ring with up to two groups selected from halo, (C1-C3)alkyl, (C1-C3)alkoxy, or CF3, . -NR5R6 wherein
R5 is H or (C1-C3)alkyl, and
R6 is (C1-C3)alkyl optionally substituted with
• indoly-2-yl optionally substituted with halo,
• phenyl optionally substituted with up to two groups selected from (C1-C3)alkoxy, or
• benzodioxan-6-yl, or
• OR7 wherein R7 is
• H,
• (C3-C6)cycloalkyl optionally substituted with (C1-C3)alkyl,
• 2,3-dihydro-inden-2-yl,
• tetrahydrofuran-yl, or
• (C1-C6)alkyl optionally substituted with
• F up to the perfluoro level,
• (C3-C6)cycloalkyl,
• (C1-C3)alkoxy,
• — N[(C1-C3)alkyl]2,
• — NH[(C1-C3)alkyl],
• morpholin-1-yl, • phenyl optionally substituted with up to two groups selected from CF3, halo, (C1-C3)alkyl, NO2, (C1-C3)alkoxy, or N[(C1-C3)alkyl]2,
• 2,3-dihydro-benzo[1 ,4]dioxin-2-yl,
• 2,3-dihydro-1 H-indol-2-yl,
• tetrahydrofuran-yl,
• 2-oxo-2,3-dihydro-benzoimidazol-1 -yl,
• 2-oxo-pyrrolidin-1-yl, or
• — N[(C1-C3)alkyl]phenyl, wherein the phenyl is optionally substituted with up to two groups selected from CF3, halo, (C1-C3)alkyl, (C1- C3)alkoxy, and — N[(C1-C3)alkyl]2;
R3 is H or (C1-C3)alkyl; and
R4 is H, methyl, -C(O)O-R8, or -C(O)R9; wherein
R8 is (C1-C4)alkyl or phenyl optionally substituted with halo, (C1-C3)alkyl, (C1-C3)alkoxy, or CF3, and
R9 is (C1-C4)alkyl or phenyl optionally substituted with halo, (C1-C3)alkyl, (C1-C3)alkoxy, or CF3.
3. The compound of claim 1 , wherein
R1 is
• (C1-C4)alkyl, or
• (C3-C6)cycloalkyl optionally substituted with (C1-C3)alkyl; R2 is
• phenylpyrrolidin-1-yl optionally substituted on the phenyl ring with up to two groups selected from halo, (Ci-C3)alkyl, (C1-C3)alkoxy, or CF3,
• -NR5R6 wherein
R5 is H or (C1-C3)alkyl, and
R6 is (C1-C3)alkyl optionally substituted with
• indoly-2-yl optionally substituted with halo,
• phenyl optionally substituted with up to two groups selected from (C1-C3)alkoxy, or
• benzodioxan-6-yl, or
• OR7 wherein R7 is
• (C3-C6)cycloalkyl optionally substituted with (C1-C3)alkyl, or
• (C1-C6)alkyl optionally substituted with
• F up to the perfluoro level,
• (C3-C6)cycloalkyl,
• (C1-C3)alkoxy,
• phenyl optionally substituted with up to two groups selected from CF3, halo, (C1-C3)alkyl, NO2, (C1-C3)alkoxy, or N[(C1-C3)alkyl]2;
R3 is H or (C1-C3)alkyl; and R4 is H, methyl, -C(O)O-R8, or -C(O)R9; wherein
R8 is (C1-C4)alkyl or phenyl optionally substituted with halo, (C1-C3)alkyl, (C1-C3)alkoxy, or CF3, and
R9 is (C1-C4)alkyl or phenyl optionally substituted with halo, (C1-C3)alkyl, (C1-C3)alkoxy, or CF3.
4. The compound of claim 1 , wherein
R1 is (C3-C6)cycloalkyl optionally substituted with (C1-C3)alkyl;
R2 is
• phenylpyrrolidin-1-yl optionally substituted on the phenyl ring with up to two groups selected from halo, (C1-C3)alkyl, (C1-C3)aIkoxy, or CF3, or
• OR7 wherein R7 is
• (C3-C6)cycloalkyl optionally substituted with (C1-C3)alkyl, or
• (C1-C6)alkyl optionally substituted with
• F up to the perfluoro level,
• (C3-C6)cycloalkyl,
• (C1-C3)alkoxy,
• — N[(C1-C3)alkyl]2,
• — NH[(C1-C3)alkyl],
• morpholin-1-yl,
• phenyl optionally substituted with up to two groups selected from CF3, halo, (C1-C3)alkyl, NO2, (C1-C3)alkoxy, or N[(C1-C3)alkyl]2l
• 2,3-dihydro-benzo[1 ,4]dioxin-2-yl,
• 2,3-dihydro-1 H-indol-2-yl,
• tetrahydrofuran-yl,
• 2-0X0-2, 3-dihydro-benzoimidazol-1 -yl,
• 2-oxo-pyrrolidin-1-yl, or
• — N[(C1-C3)alkyl]phenyl, wherein the phenyl is optionally substituted with up to two groups selected from CF3, halo, (C1-C3)alkyl, (C1- C3)alkoxy, and — N[(C1-C3)alkyl]2;
R3 is H or (C1-C3)alkyl; and
R4 is H, methyl.
5. The compound of claim 1 , wherein
R1 is (C3-C6)cycloalkyl optionally substituted with (C1-C3)alkyl;
R2 is
• OR7 wherein R7 is
• (C3-C6)cycloalkyl optionally substituted with (C1-C3)alkyl, or
• (C1-C6)alkyl optionally substituted with
• F up to the perfluoro level,
• (C3-C6)cycloalkyl,
• (C1-C3)alkoxy, or
• phenyl optionally substituted with up to two groups selected from CF3, halo, (C1-C3)alkyl, NO2, (C1-C3)all<oxy, or N[(CrC3)alkyl]2;
R3 is H; and
R4 is H or methyl.
6. A pharmaceutical composition comprising a therapeutically effective amount of a compound of claim 1 , or a pharmaceutically acceptable salt or ester, in combination with a pharmaceutically acceptable carrier.
7. A pharmaceutical composition comprising a therapeutically effective amount of a compound of claim 1 , or a pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable carrier and one or more pharmaceutical agents.
8. The pharmaceutical composition of claim 7, wherein said pharmaceutical agent is selected from the group consisting of PPAR ligands, insulin secretagogues, sulfonylurea drugs, α-glucosidase inhibitors, insulin sensitizers, hepatic glucose output lowering compounds, insulin and insulin derivatives, biguanides, protein tyrosine phosphatase-1 B, dipeptidyl peptidase IV, 11 beta-HSD inhibitors, anti- obesity drugs, HMG-CoA reductase inhibitors, nicotinic acid, lipid lowering drugs, ACAT inhibitors, bile acid sequestrants, bile acid reuptake inhibitors, microsomal triglyceride transport inhibitors, fibric acid derivatives, β-blockers, ACE inhibitors, calcium channel blockers, diuretics, renin inhibitors, AT-1 receptor antagonists, ET receptor antagonists, neutral endopeptidase inhibitors, vasopepsidase inhibitors, and nitrates.
9. The pharmaceutical composition of claim 7, wherein said pharmaceutical agent is selected from β-3 adrenergic receptor agonists, cannabinoid antagonists, neuropeptide-Y receptor antagonists, neuropeptide Y5 inhibitors, apo-B/MTP inhibitors, 11 β-hydroxy steroid dehydrogenase-1 inhibitors, peptide YY3.36 or analogs thereof, MCR4 agonists, CCK-A agonists, monoamine reuptake inhibitors, sympathomimetic agents, dopamine agonists, melanocyte-stimulating hormone receptor analogs, melanin concentrating hormone antagonists, leptin, leptin analogs, leptin receptor agonists, gaianin antagonists, lipase inhibitors, bombesin agonists, thyromimetic agents, dehydroepiandrosterone or analogs thereof, glucocorticoid receptor antagonists, orexin receptor antagonists, ciliary neurotrophic factor, ghrelin receptor antagonists, histamine-3 receptor antagonists, neuromedin U receptor agonists, appetite suppressants, and compound that modulates digestion and/or metabolism.
10. A method of regulating food intake comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1 or a composition of claim 6.
11. A method of treating obesity, obesity-related disorders, or eating disorders comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1 or a composition of claim 6.
12. A method of treating central nervous disorders, depressions, anxiety disorders, obsessive-compulsive disorders, sleep disorders, sexual dysfunction, psychoses, migraine, schizophrenia, drug or alcohol addiction and chronic fatigue syndrome comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1 or a composition of claim 6.
13. A method of treating diabetes or diabetes-related disorders comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1 or a pharmaceutical composition of claim 6.
14. The method of claim 13, wherein said diabetes is selected from the group consisting of type 1 diabetes, type 2 diabetes, maturity-onset diabetes of the young, latent autoimmune diabetes adult, and gestational diabetes.
15. The method of claim 13, wherein said diabetes-related disorder is selected from the group consisting of hyperglycemia, hyperinsulinemia, impaired glucose tolerance, impaired fasting glucose, dyslipidemia, hypertriglyceridemia, and insulin resistance.
16. A method of regulating food intake comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1 in combination with one or more pharmaceutical agents.
17. A method of treating obesity, obesity-related disorders, or eating disorders comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1 in combination with one or more pharmaceutical agents.
18. The method of claim 16 or 17, wherein said pharmaceutical agent is selected from β-3 adrenergic receptor agonists, cannabinoid antagonists, neuropeptide-Y receptor antagonists, neuropeptide Y5 inhibitors, apo-B/MTP inhibitors, 11 β-hydroxy steroid dehydrogenase-1 inhibitors, peptide YY3-36 or analogs thereof, MCR4 agonists, CCK-A agonists, monoamine reuptake inhibitors, sympathomimetic agents, dopamine agonists, melanocyte-stimulating hormone receptor analogs, melanin concentrating hormone antagonists, leptin, leptin analogs, leptin receptor agonists, galanin antagonists, lipase inhibitors, bombesin agonists, thyromimetic agents, dehydroepiandrosterone or analogs thereof, glucocorticoid receptor antagonists, orexin receptor antagonists, ciliary neurotrophic factor, ghrelin receptor antagonists, histamine-3 receptor antagonists, neuromedin U receptor agonists, appetite suppressants, and compound that modulates digestion and/or metabolism.
19. A method of treating central nervous disorders, depressions, anxiety disorders, obsessive-compulsive disorders, sleep disorders, sexual dysfunction, psychoses, migraine, schizophrenia, drug or alcohol addiction and chronic fatigue syndrome comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1 in combination with one or more pharmaceutical agents.
20. A method of treating diabetes or diabetes-related disorders comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1 in combination with one or more pharmaceutical agents.
21. The method of claim 20, wherein said pharmaceutical agent is selected from the group consisting of PPAR agonists, sulfonylurea drugs, non-sulfonylurea secretagogues, α-glucosidase inhibitors, insulin sensitizers, insulin secretagogues, hepatic glucose output lowering compounds, insulin, and anti-obesity agents.
22. The method of claim 20, wherein said diabetes is selected from the group consisting of type 1 diabetes, type 2 diabetes, maturity-onset diabetes of the young, latent autoimmune diabetes adult, and gestational diabetes.
23. The method of claim 20, wherein said diabetes-related disorder is selected from the group consisting of hyperglycemia, hyperinsulinemia, impaired glucose tolerance, impaired fasting glucose, dyslipidemia, hypertriglyceridemia, and insulin resistance.
24. A method of treating obesity, obesity-related disorders, eating disorders, diabetes, or diabetes-related disorders comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1 in combination with one or more agents selected from the group consisting of HMG-CoA reductase inhibitors, nicotinic acid, lipid lowering drugs, ACAT inhibitors, bile acid sequestrants, bile acid reuptake inhibitors, microsomal triglyceride transport inhibitors, fibric acid derivatives, β-blockers, ACE inhibitors, calcium channel blockers, diuretics, renin inhibitors, AT-1 receptor antagonists, ET receptor antagonists, neutral endopeptidase inhibitors, vasopepsidase inhibitors, and nitrates.
25. The method of claim 24, wherein said diabetes-related disorder is selected from the group consisting of hyperglycemia, hyperinsulinemia, impaired glucose tolerance, impaired fasting glucose, dyslipidemia, hypertriglyceridemia, and insulin resistance.
26. The method of any one of claims 16 to 25, wherein the compound of claim 1 and one or more pharmaceutical agents are administered as a single pharmaceutical dosage formulation.
27. Compounds according to claim 1 for the treatment and/or prophylaxis of obesity, obesity-related disorders, eating disorders, diabetes, diabetes-related disorders, central nervous disorders, depressions, anxiety disorders, obsessive-compulsive disorders, sleep disorders, sexual dysfunction, psychoses, migraine, schizophrenia, drug or alcohol addiction and chronic fatigue syndrome.
28. Medicament containing at least one compound according to claim 1 in combination with at least one pharmaceutically acceptable, pharmaceutically safe carrier or excipient.
29. Use of compounds according to claim 1 for manufacturing a medicament for the treatment and/or prophylaxis of obesity, obesity-related disorders, eating disorders, diabetes, diabetes-related disorders, central nervous disorders, depressions, anxiety disorders, obsessive-compulsive disorders, sleep disorders, sexual dysfunction, psychoses, migraine, schizophrenia, drug or alcohol addiction and chronic fatigue syndrome.
30. Medicaments according to claim 28 for the treatment and/or prophylaxis of obesity, obesity-related disorders, eating disorders, diabetes, diabetes-related disorders, central nervous disorders, depressions, anxiety disorders, obsessive-compulsive disorders, sleep disorders, sexual dysfunction, psychoses, migraine, schizophrenia, drug or alcohol addiction and chronic fatigue syndrome.
31. A method of treating diseases or disorders associated with the 5-HT2C receptor comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1 or a composition of claim 6.
32. A method of treating diseases or disorders associated with the 5-HT2C receptor comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1 in combination with one or more pharmaceutical agents.
PCT/US2005/037193 2004-10-15 2005-10-14 Tetrahydro-5h-pyrimido[4,5-d]azepine derivatives useful for the treatment of diseases associated with the 5-ht2c receptor WO2006044762A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US61919504P 2004-10-15 2004-10-15
US60/619,195 2004-10-15

Publications (2)

Publication Number Publication Date
WO2006044762A2 true WO2006044762A2 (en) 2006-04-27
WO2006044762A3 WO2006044762A3 (en) 2006-08-03

Family

ID=36203607

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/037193 WO2006044762A2 (en) 2004-10-15 2005-10-14 Tetrahydro-5h-pyrimido[4,5-d]azepine derivatives useful for the treatment of diseases associated with the 5-ht2c receptor

Country Status (1)

Country Link
WO (1) WO2006044762A2 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007109355A3 (en) * 2006-03-21 2007-11-15 Janssen Pharmaceutica Nv 6,7,8, 9 -tetrahydro- 5h- pyrimido [4, 5-d] azepin-4-yl] -amine derivatives as modulators of trpvl for the treatment of pain
WO2007132841A1 (en) 2006-05-16 2007-11-22 Takeda Pharmaceutical Company Limited Fused heterocyclic compound and use thereof
WO2007140213A1 (en) * 2006-05-26 2007-12-06 Forest Laboratories Holdings Limited Pyridoazepine derivatives
WO2008117169A1 (en) * 2007-03-23 2008-10-02 Pfizer Limited Pyrimido [4, 5-d] azepine derivatives as 5-ht2c agonists
WO2009063992A1 (en) 2007-11-15 2009-05-22 Takeda Pharmaceutical Company Limited Condensed pyridine derivative and use thereof
WO2012030953A1 (en) 2010-09-01 2012-03-08 Arena Pharmaceuticals, Inc. 5-ht2c receptor agonists in the treatment of disorders ameliorated by reduction of norepinephrine level
US8637527B2 (en) 2007-12-17 2014-01-28 Janssen Pharmaceutica Nv Imidazolo-, oxazolo-, and thiazolopyrimidine modulators of TRPV1
WO2015066344A1 (en) 2013-11-01 2015-05-07 Arena Pharmaceuticals, Inc. 5-ht2c receptor agonists and compositions and methods of use
WO2019131902A1 (en) 2017-12-27 2019-07-04 武田薬品工業株式会社 Therapeutic agent for stress urinary incontinence and fecal incontinence
WO2020039093A1 (en) * 2018-08-24 2020-02-27 Jaguahr Therapeutics Pte Ltd Tetrahydropyridopyrimidine derivatives as ahr modulators
WO2021173082A1 (en) * 2020-02-26 2021-09-02 Jaguahr Therapeutics Pte Ltd Pyridopyrimidine derivatives useful in modulation of ahr signalling

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995021844A1 (en) * 1994-02-10 1995-08-17 Smithkline Beecham Plc 5ht2b receptor antagonists condensed indoles
US20030225057A1 (en) * 2002-04-12 2003-12-04 Arena Pharmaceuticals, Inc. 5HT2c receptor modulators

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995021844A1 (en) * 1994-02-10 1995-08-17 Smithkline Beecham Plc 5ht2b receptor antagonists condensed indoles
US20030225057A1 (en) * 2002-04-12 2003-12-04 Arena Pharmaceuticals, Inc. 5HT2c receptor modulators

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9422293B2 (en) 2006-03-21 2016-08-23 Janssen Pharmaceutica Nv Tetrahydro-pyrimidoazepines as modulators of TRPV1
EA017069B1 (en) * 2006-03-21 2012-09-28 Янссен Фармацевтика Н.В. Tetrahydropyrimidoazepines as modulators of trpv1
JP2009530396A (en) * 2006-03-21 2009-08-27 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ Tetrahydro-pyrimidoazepine as a modulator of TRPV1
WO2007109355A3 (en) * 2006-03-21 2007-11-15 Janssen Pharmaceutica Nv 6,7,8, 9 -tetrahydro- 5h- pyrimido [4, 5-d] azepin-4-yl] -amine derivatives as modulators of trpvl for the treatment of pain
US9738649B2 (en) 2006-03-21 2017-08-22 Janssen Pharmaceutica N.V. Tetrahydro-pyrimidoazepines as modulators of TRPV1
EP2727585A1 (en) 2006-05-16 2014-05-07 Takeda Pharmaceutical Company Limited In-vivo screening method
WO2007132841A1 (en) 2006-05-16 2007-11-22 Takeda Pharmaceutical Company Limited Fused heterocyclic compound and use thereof
EP2742936A1 (en) 2006-05-16 2014-06-18 Takeda Pharmaceutical Company Limited Fused heterocyclic compound and use thereof
WO2007140213A1 (en) * 2006-05-26 2007-12-06 Forest Laboratories Holdings Limited Pyridoazepine derivatives
US7897595B2 (en) 2006-05-26 2011-03-01 Forest Laboratories Holdings Limited Pyridoazepine derivatives
US7928099B2 (en) 2007-03-23 2011-04-19 Pfizer Inc Pyrimido [4,5-D] azepine derivatives as 5-HT2c agonists
WO2008117169A1 (en) * 2007-03-23 2008-10-02 Pfizer Limited Pyrimido [4, 5-d] azepine derivatives as 5-ht2c agonists
WO2009063992A1 (en) 2007-11-15 2009-05-22 Takeda Pharmaceutical Company Limited Condensed pyridine derivative and use thereof
EP2789338A2 (en) 2007-11-15 2014-10-15 Takeda Pharmaceutical Company Limited Condensed pyridine derivate and use thereof
US9440978B2 (en) 2007-12-17 2016-09-13 Janssen Pharmaceutica Nv Imidazolo-, oxazolo-, and thiazolopyrimidine modulators of TRPV1
US8637527B2 (en) 2007-12-17 2014-01-28 Janssen Pharmaceutica Nv Imidazolo-, oxazolo-, and thiazolopyrimidine modulators of TRPV1
WO2012030953A1 (en) 2010-09-01 2012-03-08 Arena Pharmaceuticals, Inc. 5-ht2c receptor agonists in the treatment of disorders ameliorated by reduction of norepinephrine level
WO2015066344A1 (en) 2013-11-01 2015-05-07 Arena Pharmaceuticals, Inc. 5-ht2c receptor agonists and compositions and methods of use
WO2019131902A1 (en) 2017-12-27 2019-07-04 武田薬品工業株式会社 Therapeutic agent for stress urinary incontinence and fecal incontinence
JP2021535213A (en) * 2018-08-24 2021-12-16 ジャガー セラピューティクス ピーティーイーリミテッド Tetrahydropyridopyrimidine derivative as an AhR modulator
WO2020039093A1 (en) * 2018-08-24 2020-02-27 Jaguahr Therapeutics Pte Ltd Tetrahydropyridopyrimidine derivatives as ahr modulators
CN112739698A (en) * 2018-08-24 2021-04-30 捷豹治疗有限公司 Tetrahydropyrimidine derivatives as AHR modulators
WO2021173082A1 (en) * 2020-02-26 2021-09-02 Jaguahr Therapeutics Pte Ltd Pyridopyrimidine derivatives useful in modulation of ahr signalling

Also Published As

Publication number Publication date
WO2006044762A3 (en) 2006-08-03

Similar Documents

Publication Publication Date Title
WO2006044762A2 (en) Tetrahydro-5h-pyrimido[4,5-d]azepine derivatives useful for the treatment of diseases associated with the 5-ht2c receptor
CA3140972A1 (en) Combinations comprising benzodioxol as glp-1r agonists for use in the treatment of nash/nafld and related diseases
JP2023524493A (en) heterocyclic GLP-1 agonists
EP1720863A2 (en) Heteroarylaminopyrazole derivatives useful for the treatment of diabetes
CA2926568C (en) Antagonists of prostaglandin ep3 receptor
JP2010520199A (en) Piperidine derivatives and methods of use thereof
EP3681857B1 (en) Fluorophenyl beta-hydroxyethylamines and their use in the treatment of hyperglycaemia
EA032356B1 (en) Diacylglycerol acyltransferase 2 inhibitors for use in the treatment of metabolic and related disorders
WO2008040974A1 (en) Indoles for use as dpp-iv inhibitors
JP2009541463A (en) Piperidine or pyrrolidine urea derivatives, their preparation and their therapeutic use
AU2015245260A1 (en) 2-amino-6-methyl-4,4a,5,6-tetrahydropyrano[3,4-d][1,3]thiazin-8a(8H)-yl-1,3-thiazol-4-yl amides
TW201726688A (en) N-[2-(2-amino-6,6-disubstituted-4,4a,5,6-tetrahydropyrano[3,4-d][1,3] thiazin-8a(8H)-yl)-1,3-thiazol-4-yl] amides
KR20200100111A (en) Triazolobenzazepine as vasopressin V1A receptor antagonist
WO2005112923A2 (en) 5-anilino-4-heteroarylpyrazole derivatives useful for the treatment of diabetes
EP2865666B1 (en) Hexahydropentaleno derivatives, preparation method and use in medicine thereof
EP3240783A1 (en) New benzimidazole derivatives as antihistamine agents
WO2003057213A2 (en) Cyclohexano- and cycloheptapyrazole derivative compounds, for use in diseases associated with the 5-ht2c receptor
EP1513831A1 (en) Novel compounds, their use and preparation
TW201726671A (en) N-[2-(3-amino-2,5-dimethyl-1,1-dioxido-5,6-dihydro-2H-1,2,4-thiadiazin-5-yl)-1,3-thiazol-4-yl] amides
EP1613318A2 (en) Compounds and their use to treat diabetes and related disorders
EP0973766B1 (en) Quinoxalinediones
US6333326B1 (en) Quinoxalinediones
KR101771794B1 (en) Triazolo-arylacrylonitrile derivatives and use thereof
US20060094714A1 (en) Compounds and their use to treat diabetes and related disorders
JP2023519605A (en) Compounds active against nuclear receptors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 05815538

Country of ref document: EP

Kind code of ref document: A2